Molecular and Cel Signaling
Molecular and Cel Signaling
Molecular and Cel Signaling
PHYSICS
BIOMEDICAL ENGINEERING
Martin Beckerman
Molecular and
Cellular Signaling
With 227 Figures
Martin Beckerman
Y12 National Security Complex
Oak Ridge, TN 37831-7615
USA
[email protected]
SPIN 10948309
(BS/EB)
Series Preface
The elds of biological and medical physics and biomedical engineering are
broad, multidisciplinary, and dynamic. They lie at the crossroads of frontier
research in physics, biology, chemistry, and medicine. The Biological and
Medical Physics/Biomedical Engineering series is intended to be comprehensive, covering a broad range of topics important to the study of the
physical, chemical, and biological sciences. Its goal is to provide scientists
and engineers with textbooks, monographs, and reference works to address
the growing need for information.
Books in the series emphasize established and emergent areas of science
including molecular, membrane, and mathematical biophysics; photosynthetic energy harvesting and conversion; information processing; physical
principles of genetics; sensory communications; and automata networks,
neural networks, and cellular automata. Equally important will be coverage
of applied aspects of biological and medical physics and biomedical engineering, such as molecular electronic components and devices, biosensors,
medicine, imaging, physical principles of renewable energy production,
advanced prostheses, and environmental control and engineering.
Oak Ridge, Tennessee
Elias Greenbaum
Series Editor-in-Chief
Preface
viii
Preface
ourselves. The genomes of worms, ies, mice, and humans are roughly
comparable, and only a factor of two or three larger than those of some
bacteria. The good engineering of biological systems is exemplied by the
above-mentioned partition of cellular processes into the xed infrastructure and the control layer. This makes possible machinery that always works
the same way in any cell at any time, and whose interactions can be exactly
known, while allowing for the machinerys regulation by the variable
control layer at well-dened control points.
Another example of good engineering design is that of modularity of
design. Proteins, especially signaling proteins, are modular in design and
their components can be transferred, arranged, and rearranged to make
many different proteins. The protein components interact with one another
through their interfaces. There are interfaces for interactions with other
proteins and with lipids DNA and RNA. Modularity is encountered not
only in the largely independent components, but also in the DNA regulatory sequences. These sequences serve as control points for the networks
that regulate gene expression. The DNA regulatory sequences can also
be rearranged in a multitude of ways along the chromosomes, and these
rearrangements, rather than the genes themselves, are largely responsible
for the richness of life on Earth. Two of the key objectives of the text are
to examine how modularity in design is used and how interfaces are
exploited. X-ray crystal structures and nuclear magnetic resonance (NMR)
solution structures provide insights at the atomic level of how the interfaces
between modules operate, and these will be looked at throughout the text.
One of the great conceptual breakthroughs in explorations of the control
layer was the idea that signaling proteins involved in cell-to-cell communication are organized into signaling pathways. In a signaling pathway, there
is a starting point, usually a receptor at the plasma membrane, and an endpoint (control point), more often than not a transcription regulatory site
in the nucleus, and there is a linear route leading from one to the other.
In spite of the enormous complexity of metazoans, there are only about a
dozen or so such pathways. These will be explored in the context of where
they are most strongly associated. For example, some pathways are prominent during development and are best understood in that context. Other
pathways are associated with stress responses and are best understood
within that framework, and still others are associated with immune
responses.
Signaling and the cellular responses to signals are complex.The responses
are controlled by a plethora of positive and negative feedback loops. The
presence of feedback complicates the simple picture of a linear pathway,
but this aspect is an essential part of the signaling process. Positive feedback ensures that once the appropriate thresholds are passed there will be
a rm commitment to a specic action and the system will not jump back
and forth between alternative responses. Negative feedback generates the
thresholds that ensure random excursions and perturbations do not unnec-
Preface
ix
essarily commit the cell to some irreversible response when it ought not to,
and it permits the cells to turn off the signaling once it has served its
purpose. These feedback loops will be examined along with the discussions
of the linear signaling pathways.
The goal of this textbook is to provide an introduction to the molecular
and cellular signaling processing comprising the control layer. The topic is
a vast one, and it is not possible to cover every possible aspect and still keep
the text concise and readable. To achieve the stated goal, material of a historical nature has been omitted, as have lengthy descriptions of all proteins
identied as being involved in the particular aspect of signaling being considered. In place of such an encyclopedic approach, selected processes are
presented step-by-step from start to end. These examples serve as simple
models of how the control process is carried out.
Oak Ridge, Tennessee
Martin Beckerman
Contents
Series Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Guide to Acronyms . . . . . . . . . . . . . . . . . . . . . . . . . . .
1.
2.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . .
1.1
Prokaryotes and Eukaryotes . . . . . . . . . . . . . .
1.2
The Cytoskeleton and Extracellular Matrix . . . . .
1.3
Core Cellular Functions in Organelles . . . . . . . .
1.4
Metabolic Processes in Mitochondria and
Chloroplasts . . . . . . . . . . . . . . . . . . . . . . .
1.5
Cellular DNA to Chromatin . . . . . . . . . . . . . .
1.6
Protein Activities in the Endoplasmic Reticulum and
Golgi Apparatus . . . . . . . . . . . . . . . . . . . . .
1.7
Digestion and Recycling of Macromolecules . . . . .
1.8
Genomes of Bacteria Reveal Importance of
Signaling . . . . . . . . . . . . . . . . . . . . . . . . .
1.9
Organization and Signaling of Eukaryotic Cell . . .
1.10 Fixed Infrastructure and the Control Layer . . . . .
1.11 Eukaryotic Gene and Protein Regulation . . . . . .
1.12 Signaling Malfunction Central to Human
Disease . . . . . . . . . . . . . . . . . . . . . . . . . .
1.13
Organization of Text . . . . . . . . . . . . . . . . . .
The Control Layer . . . . . . . . . . . . . . . . . . . . . . .
2.1
Eukaryotic Chromosomes Are Built from
Nucleosomes . . . . . . . . . . . . . . . . . . . . . .
2.2
The Highly Organized Interphase Nucleus . . . . .
2.3
Covalent Bonds Dene the Primary Structure of a
Protein . . . . . . . . . . . . . . . . . . . . . . . . .
2.4
Hydrogen Bonds Shape the Secondary Structure .
2.5
Structural Motifs and Domain Folds:
Semi-Independent Protein Modules . . . . . . . .
v
vii
xxv
.
.
.
.
1
1
2
3
.
.
4
5
.
.
6
8
.
.
.
.
9
10
12
13
.
.
15
16
. .
21
. .
. .
22
23
. .
. .
26
27
. .
29
xi
xii
Contents
2.6
2.7
2.8
2.9
2.10
2.11
2.12
2.13
2.14
2.15
2.16
. . .
29
. . .
30
. . .
32
. . .
33
. . .
. . .
34
36
. . .
36
. . .
37
. . .
38
. . .
. . .
39
40
. .
45
. .
. .
. .
46
49
49
. .
. .
53
53
. .
56
. .
. .
57
58
. .
60
. .
61
. .
61
. .
. .
. .
62
63
64
Contents
4.
5.
Macromolecular Forces . . . . . . . . . . . . . . . . . . .
4.1
Amino Acids Vary in Size and Shape . . . . . . .
4.2
Amino Acids Behavior in Aqueous
Environments . . . . . . . . . . . . . . . . . . . .
4.3
Formation of H-Bonded Atom Networks . . . .
4.4
Forces that Stabilize Proteins . . . . . . . . . . .
4.5
Atomic Radii of Macromolecular Forces . . . . .
4.6
Osmophobic Forces Stabilize Stressed Cells . . .
4.7
Protein Interfaces Aid Intra- and Intermolecular
Communication . . . . . . . . . . . . . . . . . . .
4.8
Interfaces Utilize Shape and Electrostatic
Complementarity . . . . . . . . . . . . . . . . . .
4.9
Macromolecular Forces Hold Macromolecules
Together . . . . . . . . . . . . . . . . . . . . . . .
4.10 Motion Models of Covalently Bonded Atoms . .
4.11
Modeling van der Waals Forces . . . . . . . . . .
4.12 Molecular Dynamics in the Study of System
Evolution . . . . . . . . . . . . . . . . . . . . . . .
4.13 Importance of Water Molecules in Cellular
Function . . . . . . . . . . . . . . . . . . . . . . .
4.14 Essential Nature of Protein Dynamics . . . . . .
xiii
. . .
. . .
71
71
.
.
.
.
.
.
.
.
.
.
72
74
74
75
76
. . .
77
. . .
78
. . .
. . .
. . .
79
79
81
. . .
83
. . .
. . .
84
85
.
.
.
.
.
. .
89
. .
. .
90
91
. .
92
. .
. .
93
94
.
.
.
.
.
.
.
.
96
98
99
100
. .
. .
101
102
. .
103
. .
104
xiv
Contents
7.
.
.
111
112
113
.
.
115
116
.
.
.
117
118
119
121
.
.
122
123
123
.
.
124
125
125
.
.
128
129
.
.
.
131
133
134
. . . . .
. . . . .
. . . . .
139
140
141
. . . . .
. . . . .
143
144
. . . . .
145
. . . . .
146
. . . . .
. . . . .
147
148
Contents
7.9
7.10
7.11
7.12
7.13
7.14
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
149
150
152
154
154
. . . . .
156
xv
.
.
.
161
162
163
165
.
.
.
.
.
.
165
167
168
169
170
171
172
.
.
173
174
176
177
178
179
180
182
. . . . . . . .
. . . . . . . .
187
188
. . . . . . . .
190
. . . . . . . .
192
xvi
Contents
9.4
9.5
9.6
9.7
9.8
9.9
9.10
9.11
9.12
9.13
9.14
9.15
9.16
9.17
9.18
9.19
9.20
10.
. . .
193
. . .
194
. . .
. . .
196
196
. . .
198
.
.
.
.
.
.
.
.
199
200
202
203
. . .
205
. . .
. . .
. . .
206
207
208
. . .
209
. . .
. . .
211
212
. . .
213
. . . .
221
. . . .
. . . .
. . . .
221
223
224
. . . .
225
. . . .
226
. . . .
228
. . . .
229
. . . .
230
. . . .
231
.
.
.
.
Contents
xvii
. .
232
. .
. .
233
234
. .
. .
235
236
. .
237
. .
239
. .
239
247
248
249
250
251
252
254
256
256
258
259
261
262
263
264
266
267
268
xviii
Contents
. .
. .
275
276
. .
. .
. .
278
279
280
. .
281
. .
282
. .
. .
. .
284
285
288
. .
. .
289
291
. .
292
. .
292
. .
. .
. .
293
295
297
. .
297
. .
299
.
.
.
305
306
307
.
.
308
311
313
.
.
.
.
314
317
317
318
Contents
Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
14.1 Several Critical Mutations Generate a
Transformed Cell . . . . . . . . . . . . . . . . . . . .
14.2 Ras Switch Sticks to On Under Certain
Mutations . . . . . . . . . . . . . . . . . . . . . . . .
14.3 Crucial Regulatory Sequence Missing in Oncogenic
Forms of Src . . . . . . . . . . . . . . . . . . . . . . .
14.4 Overexpressed GFRs Spontaneously Dimerize in
Many Cancers . . . . . . . . . . . . . . . . . . . . . .
14.5 GFRs and Adhesion Molecules Cooperate to
Promote Tumor Growth . . . . . . . . . . . . . . . .
14.6 Role of Mutated Forms of Proteins in Cancer
Development . . . . . . . . . . . . . . . . . . . . . .
14.7 Translocated and Fused Genes Are Present in
Leukemias . . . . . . . . . . . . . . . . . . . . . . . .
14.8
Repair of DNA Damage . . . . . . . . . . . . . . . .
14.9 Double-Strand-Break Repair Machinery . . . . . . .
14.10 How Breast Cancer (BRCA) Proteins Interact with
DNA . . . . . . . . . . . . . . . . . . . . . . . . . . .
14.11 PI3K Superfamily Members that Recognize
Double-Strand Breaks . . . . . . . . . . . . . . . . .
14.12 Checkpoints Regulate Transition Events in a
Network . . . . . . . . . . . . . . . . . . . . . . . . .
14.13 Cyclin-Dependent Kinases Form the Core of
Cell-Cycle Control System . . . . . . . . . . . . . . .
14.14 pRb Regulates Cell Cycle in Response to
Mitogenic Signals . . . . . . . . . . . . . . . . . . . .
14.15 p53 Halts Cell Cycle While DNA Repairs Are
Made . . . . . . . . . . . . . . . . . . . . . . . . . . .
14.16 p53 and pRb Controllers Central to Metazoan
Cancer Prevention Program . . . . . . . . . . . . . .
xix
320
.
.
321
322
323
324
325
326
331
332
334
336
336
337
338
.
.
.
339
340
342
344
345
346
347
347
349
350
xx
Contents
16.
Apoptosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
15.1 Caspases and Bcl-2 Proteins Are Key Mediators of
Apoptosis . . . . . . . . . . . . . . . . . . . . . . . .
15.2 Caspases Are Proteolytic Enzymes Synthesized as
Inactive Zymogens . . . . . . . . . . . . . . . . . . .
15.3 Caspases Are Initiators and Executioners of
Apoptosis Programs . . . . . . . . . . . . . . . . . .
15.4 There Are Three Kinds of Bcl-2 Proteins . . . . . . .
15.5
How Caspases Are Activated . . . . . . . . . . . . .
15.6 Cell-to-Cell Signals Stimulate Formation of the
DISC . . . . . . . . . . . . . . . . . . . . . . . . . . .
15.7
Death Signals Are Conveyed by the Caspase 8
Pathway . . . . . . . . . . . . . . . . . . . . . . . . .
15.8 How Pro- and Antiapoptotic Signals Are
Relayed . . . . . . . . . . . . . . . . . . . . . . . . . .
15.9 Bcl-2 Proteins Regulate Mitochondrial Membrane
Permeability . . . . . . . . . . . . . . . . . . . . . . .
15.10 Mitochondria Release Cytochrome c in Response to
Oxidative Stresses . . . . . . . . . . . . . . . . . . . .
15.11 Mitochondria Release Apoptosis-Promoting
Agents . . . . . . . . . . . . . . . . . . . . . . . . . .
15.12 Role of Apoptosome in (Mitochondrial Pathway to)
Apoptosis . . . . . . . . . . . . . . . . . . . . . . . .
15.13 Inhibitors of Apoptosis Proteins Regulate Caspase
Activity . . . . . . . . . . . . . . . . . . . . . . . . . .
15.14 Smac/DIABLO and Omi/HtrA2 Regulate IAPs . .
15.15 Feedback Loops Coordinate Actions at Various
Control Points . . . . . . . . . . . . . . . . . . . . . .
15.16 Cells Can Produce Several Different Kinds of
Calcium Signals . . . . . . . . . . . . . . . . . . . . .
15.17 Excessive [Ca2+] in Mitochondria Can Trigger
Apoptosis . . . . . . . . . . . . . . . . . . . . . . . .
15.18 p53 Promotes Cell Death in Response to Irreparable
DNA Damage . . . . . . . . . . . . . . . . . . . . . .
15.19 Anti-Cancer Drugs Target the Cells Apoptosis
Machinery . . . . . . . . . . . . . . . . . . . . . . . .
Gene
16.1
16.2
16.3
Regulation in Eukaryotes . . . . . . . . . . . .
Organization of the Gene Regulatory Region
How Promoters Regulate Genes . . . . . . .
TFs Bind DNA Through Their DNA-Binding
Domains . . . . . . . . . . . . . . . . . . . . .
352
354
359
360
361
.
.
.
362
363
365
366
367
368
369
371
372
373
.
.
374
375
375
376
377
378
379
. . . . .
. . . . .
. . . . .
385
386
387
. . . . .
389
Contents
16.4
16.5
16.6
16.7
16.8
16.9
16.10
16.11
16.12
16.13
16.14
16.15
16.16
16.17
17.
xxi
. . .
392
. . .
393
. . .
393
. . .
394
. . .
395
. . .
397
. . .
398
. . .
399
. . .
. . .
. . .
400
401
403
. . .
404
. . .
. . .
406
407
. .
411
. .
412
. .
412
. .
414
. .
. .
. .
414
416
417
. .
418
. .
. .
419
421
. .
422
. .
424
xxii
Contents
Regulation by Viruses . . . . . . . . . . . . . . . . . . . . . .
18.1 How Viruses Enter Their Host Cells . . . . . . . . .
18.2 Viruses Enter and Exit the Nucleus in
Several Ways . . . . . . . . . . . . . . . . . . . . . . .
18.3
Ways that Viruses Exit a Cell . . . . . . . . . . . . .
18.4 Viruses Produce a Variety of Disorders in
Humans . . . . . . . . . . . . . . . . . . . . . . . . .
18.5 VirusHost Interactions Underlie Virus Survival and
Proliferation . . . . . . . . . . . . . . . . . . . . . . .
18.6 Multilayered Defenses Are Balanced by
Multilayered Attacks . . . . . . . . . . . . . . . . . .
18.7 Viruses Target TNF Family of Cytokines . . . . . . .
18.8 Hepatitis C Virus Disables Host Cells Interferon
System . . . . . . . . . . . . . . . . . . . . . . . . . .
18.9 Human T Lymphotropic Virus Type 1 Can Cause
Cancer . . . . . . . . . . . . . . . . . . . . . . . . . .
18.10 DNA and RNA Viruses that Can Cause Cancer . .
18.11 HIV Is a Retrovirus . . . . . . . . . . . . . . . . . . .
18.12 Role of gp120 Envelope Protein in HIV . . . . . . .
18.13 Early-Acting tat, rev, and nef Regulatory
Genes . . . . . . . . . . . . . . . . . . . . . . . . . . .
18.14 Late-Acting vpr, vif, vpu, and vpx Regulatory
Genes . . . . . . . . . . . . . . . . . . . . . . . . . . .
18.15 Bacteriophages Two Lifestyles: Lytic and
Lysogenic . . . . . . . . . . . . . . . . . . . . . . . . .
18.16 Deciding Between Lytic and Lysogenic Lifestyles . .
18.17 Encoding of Shiga Toxin in E. coli . . . . . . . . . .
426
426
428
.
.
.
.
430
430
431
433
434
435
436
.
.
441
442
.
.
442
443
444
445
.
.
446
447
447
.
.
.
.
449
450
452
453
454
456
.
.
.
457
458
459
Contents
19.
20.
21.
xxiii
Ion Channels . . . . . . . . . . . . . . . . . . . . . . . . . . . .
19.1
How Membrane Potentials Arise . . . . . . . . . . . .
19.2 Membrane and Action Potentials Have Regenerative
Properties . . . . . . . . . . . . . . . . . . . . . . . . .
19.3 HodgkinHuxley Equations Describe How Action
Potentials Arise . . . . . . . . . . . . . . . . . . . . . .
19.4 Ion Channels Have Gates that Open and Close . . . .
19.5 Families of Ion Channels Expressed in Plasma
Membrane of Neurons . . . . . . . . . . . . . . . . . .
19.6
Assembly of Ion Channels . . . . . . . . . . . . . . . .
19.7 Design and Function of Ion Channels . . . . . . . . .
19.8 Gates and Filters in Potassium Channels . . . . . . . .
19.9 Voltage-Gated Chloride Channels Form a
Double-Barreled Pore . . . . . . . . . . . . . . . . . .
19.10 Nicotinic Acetylcholine Receptors Are Ligand-Gated
Ion Channels . . . . . . . . . . . . . . . . . . . . . . . .
19.11 Operation of Glutamate Receptor Ion Channels . . .
465
466
Neural Rhythms . . . . . . . . . . . . . . . . . . . . . . . . . .
20.1 Heartbeat Is Generated by Pacemaker Cells . . . . .
20.2 HCN Channels Role in Pacemaker Activities . . . . .
20.3 Synchronous Activity in the Central Nervous
System . . . . . . . . . . . . . . . . . . . . . . . . . . .
20.4
Role of Low Voltage-Activated Calcium Channels . . . .
20.5 Neuromodulators Modify the Activities of
Voltage-Gated Ion Channels . . . . . . . . . . . . . . .
20.6 Gap Junctions Formed by Connexins Mediate
Rapid Signaling Between Cells . . . . . . . . . . . . .
20.7 Synchronization of Neural Firing . . . . . . . . . . . .
20.8 How Spindling Patterns Are Generated . . . . . . . .
20.9 Epileptic Seizures and Abnormal Brain Rhythms . . .
20.10 Swimming and Digestive Rhythms in Lower
Vertebrates . . . . . . . . . . . . . . . . . . . . . . . . .
20.11 CPGs Have a Number of Common Features . . . . .
20.12 Neural Circuits Are Connected to Other Circuits and
Form Systems . . . . . . . . . . . . . . . . . . . . . . .
20.13 A Variety of Neuromodulators Regulate Operation
of the Crustacean STG . . . . . . . . . . . . . . . . . .
20.14 Motor Systems Adapt to Their Environment and
Learn . . . . . . . . . . . . . . . . . . . . . . . . . . . .
487
487
489
468
470
472
474
476
478
478
479
480
483
492
492
494
495
497
498
498
499
502
504
505
506
511
512
514
xxiv
Contents
21.3
515
518
520
521
.
.
523
524
525
.
.
.
.
526
528
529
529
531
532
533
Glossary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
539
Index
553
21.4
21.5
21.6
21.7
21.8
21.9
21.10
21.11
21.12
21.13
21.14
21.15
21.16
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Guide to Acronyms
5-hydroxytryptamine (serotonin)
AA
AC
ACE
ACF
ACh
ACTH
ADAM
ADHD
ADP
AFM
AGC
AHL
AIDS
AIF
AIP
AKAP
ALK
ALS
arachidonic acid
adenylyl cyclase
angiotensin-converting enzyme
ATP-dependent chromatin assembly and remodeling factor
acetylcholine
adrenocorticotropic hormone
a disintegrin and metalloprotease
attention-decit hyperactivity disorder
adenosine diphosphate
atomic force microscopy
PKA, PKG, PKC family
acetyl homoserine lactase
acquired immunodeciency syndrome
apoptosis inducing factor
autoinducing peptides
A-kinase anchoring protein
activin receptor-like kinase
amytrophic lateral sclerosis
xxv
xxvi
Guide to Acronyms
AMP
AMPA
AMPK
ANT
APC
APC
APP
ARC-L
Arf
ARF
ARR
ATM
ATP
ATR
AVN
AVP
adenosine monophosphate
a-amino-3-hydroxyl-5-methyl-4-isoxazole propionate acid
AMP-dependent protein kinase
adenosine nucleotide translocator
adenomatous polyposis coli
antigen-presenting cell
amyloid b protein precursor
activation-recruited coactivator-large
ADP-ribosylation factor
alternative reading frame (of exon 2)
Arabidopsis response regulator
ataxia-telangeictasia mutated
adenosine triphosphate
ATM and Rad3-related
atrioventricular node
vasopressin
Bcl-2
BCR
BDNF
BER
BFGF
BIR
BLV
BMP
BRCA1
BRCT
BRE
bZIP
B cell leukemia 2
B-cell receptor
brain-derived neurotrophic factor
base excision repair
basic broblast growth factor
baculoviral IAP repeat
bovine leukemia virus
bone morphogenetic protein
breast cancer 1
BRCA1 C-terminal
TFIIB recognition element
basic region leucine zipper
C1
CAD
CaM
CaMKII
cAMP
CAP
CAPRI
CaR
CARD
CASK
CB
CBP
CBP
CD
Cdc25
Cdk
Guide to Acronyms
cDNA
CFP
CFTR
cGMP
CHRAC
Chromo
Ci
Ck2
ClC
Clk
CMGC
CNG
CNS
CNTF
CoA
COX
CPG
CR
CRD
CRE
CREB
CRF
CRH
CRSP
CSF
cSMAC
CST
complementary DNA
cyan uorescent protein
cystic brosis transmembrane conductance regulator
cyclic guanosine monophosphate
chromatin accessibility complex
chromatin organization modier
cubitus interruptus
casein kinase-2
chloride channel of the CLC family
cyclin-dependent kinase-like kinase
CDK, MAPK, GSK-3 CLK, CK2
cyclic nucleotide-gated
central nervous system
ciliary neurotrophic factor
acetyl coenzyme A
cyclo-oxygenase
central pattern generator
consensus repeat
cysteine-rich domain
cAMP response element
cAMP response element-binding protein
corticotropin-releasing factor
corticotropin-releasing hormone
coactivator required for Sp1 activation
cerebrospinal uid
central supramolecular activation cluster
cortistatin
DA
DAG
DAT
dATP
DC
DCC
DD
DED
DEP
DFF
Dhh
DIABLO
DISC
DIX
DLG
DNA
DNA-PK
DPE
dopamine
diacylglycerol
dopamine transporter
deoxyadenosine triphosphate
dendritic cell
deleted in colorectal cancer
death domain
death effector domain
disheveled, egl-10, and pleckstrin
DNA fragmentation factor
desert hedgehog
direct IAP binding protein with low pI
death-inducing signaling complex
disheveled and axin
discs large
deoxyribonucleic acid
DNA-dependent protein kinase
downstream promoter element
xxvii
xxviii
Guide to Acronyms
DR
DSB
DSL
dsRNA
death receptor
double-strand break
delta/serrate/lin
double-stranded RNA
E
ECF
ECM
EEG
EGF
EGFR
eIF
EPEC
ER
ERK
ESCRT
ESE
ESI
ESS
EVH1
epinephrine (adrenaline)
extracytoplasmic function
extracellular matrix
electroencephalographic
epidermal growth factor
epidermal growth factor receptor
eukaryotic initiation factors
enteropathogenic E. coli
endoplasmic reticulum
extracellular signal-regulated kinase
endosomal-sorting complexes required for transport
exonic splice enhancer
electrospray ionization
exonic splice silencer
enabled/vasodilator-stimulated phosphoprotein homology-1
FA
FADD
FAK
FAT
FH
FHA
FNIII
FRAP
FSH
FYVE
focal adhesion
Fas-associated death domain
focal adhesion kinase
focal adhesion targeting
forkhead
forkhead associated
bronectin type III
uorescence recovery following photobleaching
follicle-stimulating hormone
Fab1p, YOTB, Vac1p, Eea1
GABA
GAP
GAS
GAS
GDI
GDNF
GDP
GEF
GFP
GFR
GH
GHIH
GHRH
g-aminobutyric acid
GTPase-activating protein
group A streptococcus
interferon-gamma activated site
GDP dissociation inhibitors
glial-derived neurotrophic factor
guanosine diphosphate
guanine nucleotide exchange factor
green uorescent protein
growth factor receptor
growth hormone
growth hormone-inhibiting hormone
growth hormone-releasing hormone
Guide to Acronyms
GIRK
GKAP
GPCR
GPI
GRH
GRIP
GRK
GSK-3
GTP
HA
HAT
HDAC
hGH
HGT
Hh
HHV
HIV
HK
HLH
HNC
hnRNP
HOG
HPt
HR
Hsp
HSV-1
hTERT
HTH
HTLV-1
hTR
HtrA2
histamine
histone acetyltransferase
histone deacetylase
human growth hormone
horizontal gene transfer
hedgehog
human herpesvirus
human immunodeciency virus
histidine kinase
helix-loop-helix
hyperpolarization-activated cyclic nucleotide gated
heterogeneous nuclear RNP
high osmolarity glycerol
histidine phosphotransfer
homologous recombination
heat shock protein
herpes simplex virus type 1
human telomerase reverse transcriptase
helix-turn-helix
human T lymphotropic virus type 1
human telomerase RNA
high temperature requirement factor A2
IAP
ICAD
ICAM
ICE
IEG
IFN
Ig
IGC
IgCAM
IGluR
IGluR
Ihh
inhibitor of apoptosis
inhibitor of CAD
intercellular cell adhesion molecule
interleukin-1b converting enzyme
immediate early gene
interferon
immunoglobulin
interchromatin granule clusters
immunoglobulin cell adhesion molecule
inhibitory glutamate receptor ion channel
ionotropic glutamate receptor
Indian hedgehog
xxix
xxx
Guide to Acronyms
IL
ILP
IN
Inr
InsP3R
IP
IPSP
IRAK
IRES
IRF
IS
IS
ISE
ISRE
ISS
ISWI
ITAM
interleukin
IAP-like protein
integrase
initiator
inositol (1,4,5) triphosphate receptor
Ischemic preconditioning
inhibitory postsynaptic potential
IL-1R-associated kinase
internal ribosomal entry site
interferon regulatory factor
immunological synapse
intracellular stores
intronic splice enhancer
interferon stimulated response element
intronic splice silencer
imitation SWI
immunoreceptor tyrosine-based activation motif
Jak
JNK
Janus kinase
c-Jun N-terminal kinase
KSHV
L
LAMP
LANA-1
LH
LNR
LNS
LPS
LRR
LTD
LTP
LTR
LZ
late (domain)
latency-associated membrane protein
latency-associated nuclear antigen type 1
luteinizing hormone
lin/notch repeat
laminin, neurexin, sex hormone-binding globulin
lipopolysaccharides
leucine-rich repeat
long-term depression
long-term potentiation
long terminal repeat
leucine zipper
MA
MAGE
MALDI
MAOI
MAP
MAPK
MCP
MD
MH1
MHC
matrix
melanoma-associated antigen
matrix-assisted laser desorption ionization
monoamine oxidase inhibitor
mitogen-activated protein
mitogen-activated protein kinase
methyl-accepting chemotaxis protein
molecular dynamics
mad homology-1
major histocompatibility complex
Guide to Acronyms
MIP
MM
MMP
MMR
MRI
mRNA
MSH
MVB
NAc
nAChR
NADE
NAIP
NBS
NC
NCAM
NE
NER
NES
NFAT
NF-kB
NGF
NH
NHEJ
NICD
NKA
NKB
NLS
NMDA
NMR
NPC
NRAGE
NRIF
NSAID
NSF
NURF
nucleus accumbens
nicotinic acetylcholine receptor
p75-associated cell death executioner
neuronal inhibitory apoptosis protein
Nijmegem breakage syndrome
nucleocapsid
neural cell adhesion molecule
norepinephrine (noradrenaline)
nucleotide excision repair
nuclear export signal (sequence)
nuclear factor of activated T cells
nuclear factor kappa B
nerve growth factor
amide (molecule)
nonhomologous end joining
notch intracellular domain
neurokinin A
neurokinin B
nuclear localization signal (sequence)
N-methyl-d-aspartate
nuclear magnetic resonance
nuclear pore complex
neurotrophin receptor-interacting MAGE homolog
neurotrophin receptor-interacting factor
nonsteroidal anti-inammatory drug
N-ethylmaleimide-sensitive fusion protein
nucleosome remodeling factor
OCT
OPR
OT
octopamine
octicopeptide repeat
oxytocin
PACAP
PAGE
PBP
PCP
PCR
xxxi
xxxii
Guide to Acronyms
PDB
PDE
PDGF
PDK
PDZ
PGHS
PH
PIC
PIH
PIKK
PIP
PKA
PKB
PKC
PKG
PKR
PLA2
PLC
PMCA
PNS
POMC
PP-II
PRH
PRL
PS
PSD
PSD-95
pSMAC
PTB
PTH
PTHrH
PTPC
PYD
QM
quantum mechanics
RACK
RAIP
RE
REM
RF
RGS
RH
RHD
RIP
Guide to Acronyms
RNA
RNP
ROS
RPA
RR
RRE
RRM
rRNA
RSC
RT
RTK
RyR
ribonucleic acid
ribonucleoprotein
reactive oxygen species
replication protein A
response regulator
rev response region
RNA recognition motif
ribosomal RNA
remodels the structure of chromatin
reverse transcriptase
receptor tyrosine kinase
ryanodine receptor
S/T
S6K
SAGA
SAM
SAM
SAN
SARA
SC1
SCR
SDS
SE
SERCA
SH2
Shh
SIV
Ski
Smac
SMCC
SN
SNAP
SNARE
SNF
SnoN
snRNA
snRNP
SODI
Sos
SP
SSRI
SST
STAT
Ste2
serine/threonine
ribosomal S6 kinase
Spt-Ada-Gen5 acetyltransferase
S-adenosyl-l-methionine
sterile a motif
sinoatrial node
smad anchor for receptor activation
Schwann cell factor-1
short consensus repeat
sodium dodecyl sulfate
spongiform encephalopathies
sarco-endoplasmic reticulum calcium ATPase
Src homology-2
sonic hedgehog
simian immunodeciency virus
SloanKettering Institute proto-oncogene
second mitochondrial activator of caspases
SRD- and MED-containing cofactor complex
sunstantia nigra
soluble NSF-attachment protein
soluble NSF-attachment protein receptor
sucrose nonfermenting
ski-related novel gene N
small nuclear RNA
small nuclear ribonucleoprotein particle
superoxide dismutase
Son-of-sevenless
substance P
selective serotonin reuptake inhibitor
somatostatin
signal transducer and activator of transcription
sterile 2
xxxiii
xxxiv
Guide to Acronyms
STG
STRE
STTK
SUMO
SWI
stomatogastric ganglion
stress responsive element
serine/threonine and tyrosine kinase
small ubiquitin-related modier
(mating type) switch
TACE
TAF
TAR
TBP
TCA
TCR
TF
TGF-b
TGIF
TM
TNF
TOF
TOP
TOR
TOS
TRADD
TRAF
TRAIL
TRAP
TRF1
tRNA
TSH
UP
UPEC
UTR
upstream (sequence)
uropathogenic E. coli
untranslated region
VAMP
VDAC
VEGF
VIP
Vps
VTA
Wg
wingless
XIAP
YFP
ZO-1
zona occludens 1
1
Introduction
Life on Earth is remarkably diverse and robust. There are organisms that
live in the deep sea and far underground, around hot midocean volcanic
vents and in cold arctic seas, and in salt brines and hot acidic springs. Some
of these creatures are methanogens that synthesize all their essential biomolecules out of H2, CO2 and salts; others are hyperthermophiles that use
H2S as a source of hydrogen and electrons, and still others are halophiles
that carry out a form of photosynthesis without chlorophyll. Some of these
extremeophiles are animallike, while others are plantlike or funguslike or
like none of these.
What is signicant about this diversity is that although the details vary
from organism to organism, all carry out the same core functions of metabolism, cell division and signaling in roughly the same manner. The underlying unity extends from tiny parasitic bacteria containing minimal
complements of genes to large differentiated multicellular plants and
animals. Each organism has a similar set of basic building blocks and
utilizes similar assembly principles. The myriad forms of life arise mostly
through rearrangements and expansions of a basic set of units rather than
different biochemistries or vastly different parts or assembly rules.
1. Introduction
matrix, and these molecules guide cellular migration and adhesion during
development. The ECM is not a simple passive medium. Instead, signaling
between ECM and the cytoskeleton is maintained throughout development
and into adult life.
The existence of a transport system in which large numbers of molecules can be moved along the cytoskeleton to and from the plasma membrane is important for signaling between cells in the body. In the immune
system, transport vacuoles move signal molecules called cytokines (antiinammatory agents such as histamines, and antimicrobial agents that
attack pathogens) to the cell surface where they are secreted from the cell.
In the nervous system, neurotransmitters are moved over long distances
down the axon and into the axon terminal via transport vesicles. In addition to outbound trafcking, there is inbound trafcking. Surface components are continually being recycled back to the internal organelles, where
they are then either reused or degraded.
Function
Respiration
Photosynthesis (plants)
Stores DNA; transcription and
splicing
Protein synthesis-translation
Processing, packaging, and shipping
Degradation and recycling
Degradation
Internalization of material
1. Introduction
Glycolysis takes place in the cytoplasm while the citric acid cycle occurs
in the mitochondrial matrix. Five complexes embedded in the inner mitochondrial membrane carry out oxidative phosphorylation (respiration). The
constituents of the ve respiratory complexesenzymes of the electron
transport chainpump protons from the matrix to the cytosol of the mitochondria, then use the free energy released by these actions to produce ATP
from adenosine diphosphate (ADP). Their photosynthetic counterparts,
photosystems I and II, function in chroloplasts.
Chloroplasts and mitochondria are enclosed in double membranes. The
inner membrane of a mitochondrion is highly convoluted, forming structures called cristae. A similar design strategy is used in chroloplasts. The
inner membrane of a chloroplast encloses a series of folded and stacked
thylakoid structures. These designs give rise to organelles possessing large
surface areas for metabolic processes. The ATP molecules are used not only
for anabolism, but also in other core cellular processes, including signaling,
where work is done and ATP is needed.
The relocation of the machinery for metabolism from the plasma membrane to internal organelles is a momentous event from the viewpoint of
signaling. It not only provides for a far greater energy supply but also frees
up a large portion of the plasma membrane for signaling. In eukaryotic cells,
the plasma membrane is studded with large numbers of signaling proteins
that are either embedded in the plasma membrane, running from the
outside to the inside, or attached to one side or the other by means of a
tether.
Figure 1.1. Genes and proteins: Depicted is the two-step process in which DNA
nucleotide sequences, or genes, are rst transcribed onto messenger RNA (mRNA)
nucleotide sequences, and then these templates are used to translate the nucleotide
sequences into amino acid sequences.
1. Introduction
1. Introduction
Figure 1.2. Movement of proteins through the endoplasmic reticulum and Golgi
apparatus: Proteins synthesis and processing start with the export of mRNAs from
the nucleus to the ribosome-studded rough endoplasmic reticulum. Nascent proteins synthesized in ribosomes are processed and then shipped in transport vesicles
to the Golgi. They pass through the cis (nearest the ER) and trans (furthest from
the ER) Golgi, and the nished products are then shipped out to their lysosomal
and the plasma membrane destinations.
10
1. Introduction
those that live under constant conditions. Because of their ability to adapt
their physiology to their environment, these bacteria may be referred to as
environmentalists. Escherichia coli and Psuedomonas aeruginosa are typical
environmentalists. Their genomes are ve to ten times larger than the
Mycoplasmas and encode ensembles of signaling and regulatory proteins
that are 50 to 100 times larger than those of the Mycoplasmas. As a result,
they are able to thrive in a variety of environmentssoil, water, airand
they deal with many different stresses. Thus, not only are the genomes
becoming larger as the bacteria become more versatile and adaptive, but
the fractions of the genomes devoted to regulatory functions are increasing as well.
Colony-forming bacteria have even larger genomes. These bacteria can
not only cope with environmental changes and stresses, but also they can
assemble into colonies and exhibit a limited form of differentiation. Their
genomes are the largest of all the prokaryotes, and the fraction of their
genomes devoted to signaling and control approaches or exceeds 10%. An
example of a prokaryote that exhibits environmental diversity and colonial
behavior is Streptomyces coelicolor. This versatile soil bacterium is used
for the production of antibiotics such as tetracycline and erythromycin. Its
signaling component accounts for 12% of its genome.
As might be expected, the genomes for multicellular plants and animals
are larger than those for even the most sophisticated prokaryotes, but not
by as much as one might expect. The rst estimates from the complete
sequencings of the human genome are in the range of 26,000 genes, of which
roughly one quarter is devoted to signaling. There are still some genes that
remain to be identied, and when further analyses are completed there may
be as many as 32,000 to 40,000 genes. This number appears to be astonishingly low. It is scarcely a factor of three larger than the genomes for some
of the bacteria. Furthermore, the genome for the bacterium S. coelicolor is
nearly as large as that for the highly differentiated, multicellular fruit y
Drosophila melanogaster.
11
Signals sent from one cell to another that allow the sender to regulate
gene expression and other cellular responses in the recipient.
Signals and sensations indicative of internal stresses and balances.
The rst category includes a diverse set of physical and chemical signals.
Most organisms can neither alter their environments nor move over large
distances. Instead, they must continually adapt their metabolism and growth
strategies to match the environmental conditions in which they nd themselves. In this grouping of signals are environmental cues important to unicellular organisms, such as light, temperature, osmolarity, pH, and nutrients.
Also included in this category are signals such as odorants and tastants
detected by sensory organs in multicellular animals, or metazoans.
The next category encompasses contact signals between surfaces and is
specic to multicellular organisms. Proteins embedded in the plasma membrane and in the ECM convey contact signals. These signals allow cells to
establish and maintain physical contact with supporting structures within
the body, and mediate two-way communication between cells in physical
contact. This category is greatly expanded in vertebrates, and includes elements of the immune system such as antibodies that have evolved from
adhesion molecules.
The third category of signals consists of the cell-to-cell messages. This
category includes pheromones, chemical signals that promote mating and
colonial behavior in unicellular organisms, and it includes the signals in
multicellular plants and animals that allow cells in tissues and organs to
work together. In humans, there are several systems of tissues, organs, and
glands that continually send and receive chemical messages. Cells of the
immune systems send and receive cytokines; cells residing in glands of the
endocrine system secrete hormones; and neurons in the nervous system
communicate using neurotransmitters and neuromodulators. Embryonic
development is controlled from cell division to cell division by programs of
gene expression. The category of cell-to-cell signals encompasses the signals
that help establish cell fate and polarity during development, and the
growth factor and hormonal signals that shape and guide the programs of
cell growth and differentiation.
The last category consists of signals generated within the cell that help
maintain proper internal balances, or homeostasis. This category includes
signals indicative of internal stresses such as improper pH conditions, excessive temperatures, and water imbalances. Macromolecules such as DNA
and proteins are marginally stable under physiological conditions. Cellular
DNA can be damaged by ultraviolet radiation, by ionizing radiation, and
by oxidative byproducts of normal cellular processes. In addition, DNA
strand breaks can occur during the DNA replication stage that precedes
mitosis and meiosis. All cells, prokaryotic and eukaryotic, possess DNA
repair systems that continually sense and repair single and double strand
breaks. In multicellular organisms, whenever DNA damage is detected and
12
1. Introduction
13
Table 1.2. Comparison of proteins in the xed infrastructure and control layer.
Property
Location
Mobility
Lifetime
Structure
Function
Fixed infrastructure
Machines/factories in organelles
Little
Longer
Fixed
Unifunctional
Control layer
Complexes in subcompartments
Considerable
Shorter
Variable
Multifunctional
Changes in how these networks are built rather than in the genes they
control underlie the increased complexity of multicellular life.
Signal proteins are not only the messengers but also the messages. Since
the messages must be conveyed from one place to another, mobility is a
key property of the proteins. Mobility is less important for functions such
as metabolism carried out as part of large molecular machines in the
organelles. Another way signal proteins differ from the other proteins is in
the presence of post-translational modications. Signal proteins are subjected to a host of post-translational modications. Some are made in the
ER and Golgi as part of the nishing process, but others, to be discussed in
the next chapter, are part of the signaling process itself. These alterations
endow the proteins with switchlike response properties, turning them on
and off.This property is absolutely essential for a signaling element, keeping
it available for conveying a message, but in an off-conguration until an
activating signal arrives.
There are several other ways that proteins belonging to the control layer
and xed infrastructure differ from one another (Table 1.2). The function
of a protein, that is, what it does, is determined by its associations, and by
where and when it establishes them. Proteins that function as part of the
transcription machinery in the nucleus or as part of the electron transport
chain in the mitochondria, usually have a single purpose, and they carry
out this task over and over. The signaling proteins form complexes too.
However, the protein complexes are smaller than the large machines used
for metabolism and replication, and the associations and interactions are
more variable. They can be different in different cell types and will even
vary somewhat over time in the same cell type. Because of this exibility,
signaling proteins are multifunctional, or pleiotropic, in their actions.
14
1. Introduction
mix of proteins being expressed at any given time in a cell. In place of DNA
regulation of prokaryotic gene and protein expression there is now a multiplicity of eukaryotic control mechanisms. Gene and protein expression can
be controlled through
DNA regulation
histone modication
splicing regulation
translation regulation
nuclear import/export
15
16
1. Introduction
17
the place where environmental conditions are sensed and most cell-to-cell
signals are received. The yeast stress and pheromone signaling systems, the
focus of Chapter 6, and the bacterial chemotaxis system, the main subject
of Chapter 7, are archetypical signaling systems. Yeasts must constantly
adapt to changing conditions in their environment, and, similarly, bacteria
must locate nutrients and sense and respond to changes in their external
environments. These systems exhibit many of the properties and principles
that characterize signaling in more complex organisms. For many years the
plasma membrane was regarded as a fairly homogeneous and passively uid
substrate into which signaling proteins were embedded. That view has
undergone considerable change over the past few years with the realization
that the plasma membrane is organized into distinct signaling domains, and
that several kinds of lipids serve as signaling intermediaries. This new
picture of the plasma membrane, along with the role of small molecules
lipid and nonlipidin signaling is explored in Chapter 8.
The immune system provides several layers of defense against viral,
bacterial, and eukaryotic pathogens. These defenses are coordinated and
regulated by networks of signaling molecules. These signaling molecules are
the subjects of Chapter 9. In response to certain signals, leukocytes, or white
blood cells, converge upon sites of an infection and kill pathogens. Chapter
10 explores the role of signaling events taking place at the cell surface that
make possible the directed movement of leukocytes into an infection site.
Leukocytes are not the only cells requiring motility. During development
cells must move about and aggregate into tissues, and nascent nerve cells
must send out growth messages to connect with other nerve cells. These
signaling processes are examined, too, in Chapter 10.
Cells secrete growth factors and hormones in order to coordinate cellular growth and proliferation. The mechanisms whereby polypeptide growth
factors are received and transduced into cellular responses are discussed in
Chapter 11. G protein-coupled receptors transduce a remarkably diverse
spectrum of messages into the cell. Among these are light, gustatory,
odorant, pheremone, pain, immunological, endocrine, and neural signals.
Signaling through these receptors is covered in Chapter 12.
During embryonic development cell-to-cell signals coordinate the genetic
programs of growth, differentiation, and proliferation. These signaling
events, along with those that regulate motility, guide the formation of organs
and tissues with well-dened boundaries composed of functionally specialized cells derived from less specialized progenitors. The determination of
which tissue or organ a particular cell becomes part of, or cell fate, is the
focus of Chapter 13.
As noted in the last section, cancer and signaling are intimately connected.
Cancer can be regarded as a collection of diseases associated with malfunctions of key elements of the control layer and DNA repair machinery.
The relationships between cancer and aberrant signaling are explored in
Chapter 14. During the past few years the subject of programmed cell death,
18
1. Introduction
General References
Alberts B, Johnson A, Lewis J, Raff M, Roberts K, and Walker P [2002]. Molecular
Biology of the Cell (4th ed). New York: Garland Publishers.
Jacob F [1977]. Evolution and tinkering. Science, 196: 11611166.
Matthews CK, van Holde KE, and Ahem KG [2000]. Biochemistry, 3rd ed. San
Francisco: Pearson Benjamin Cummings.
Stryer L [1995]. Biochemistry, 4th edition. New York: W.H. Freeman and Company.
19
Ubiquitin-Preotosome Complex
Ciechanover A [1998]. The ubiquitin-proteosome pathway: On protein death and
cell life. EMBO J., 17: 71517160.
Quality Control
Ellgaard L, Molinari M, and Helenius A [1999]. Setting the standards: Quality
control in the secretory pathway. Science, 286: 18821888.
Lindahl T, and Wood RD [1999]. Quality control by DNA repair. Science, 286:
18971905.
Wickner S, Maurizi MR, and Gottesman S [1999]. Posttranslational quality control:
Folding, refolding and degrading proteins. Science, 286: 18881893.
2
The Control Layer
21
22
23
sufciently to expose the binding sites. Steric blockages are relieved, or alternatively enhanced, by proteins that interact with and modify DNA structure,
and also by proteins that alter histone structure. In eukaryotes, the ability to
inuence gene expression by modifying histone structure adds a further
layer of control to that available through the protein-DNA binding.
Multiple control points are situated on histone tails that extend out from the
histone core beyond the DNA. As can be seen in Figure 2.2, these sites are
well exposed, thereby providing regulatory elements with ease of access.
24
division series of stages (S, G2, and M), to growth arrest (G0), or to apoptosis (cell death). Cells that do not undergo growth arrest or apoptosis enter
a synthesis (S) stage where the DNA is replicated in preparation for mitosis.
This stage is followed by a further mitosis preparatory stage (G2) where
RNAs and proteins required for mitosis are synthesized. Finally, the cell
enters into mitosis (M) where the cell divides to produce two offspring. The
three stages preceding mitosis are collectively referred to as interphase.
Chromosomes are not randomly distributed in the nucleus during interphase. Instead, they occupy distinct chromatin territories and are positioned
in a way that reects their replication status. The chromatin territories
are partitioned into ~1Mbp-chromatin domains. Interchromatin spaces
separate the chromosome territories from one another. Gene-rich early
replicating chromatin is sequestered from gene-poor chromatin, and
from mid-to-late replicating chromatin. Early replicating chromosomes are
located in the nuclear interior, while inactive and late-replicating chromo-
25
somes are situated at the periphery of the nucleus near or in contact with
the nuclear envelope.
The nuclear architecture facilitates transcription and splicing. Highly condensed chromatin serves to repress transcription while an open formation
allows access of transcription and splicing factors to the sites of transcription. Chromosomes containing active regions of transcription are more
open in their organization than those not actively undergoing transcription.
The active regions typically lie on the outer portion of the chromosome territories. They extend into the interchromatin spaces to permit a maximal
exposure of the surface of active genes to transcription and splicing factors;
the two activities, transcription and splicing, are synchronized with one
another.
The nucleus has several other kinds of structures in addition to chromatin
territories. The additional structuresnucleoli, coiled (Cajal) bodies, and
interchromatin granule clustersare nonmembrane-associated organelles.
They are used for sequestering and storage, and for preparation and assembly of materials used for gene transcription, pre-mRNA splicing, and protein synthesis. They are highly dynamic structures that form and reform
about local concentrations of materials that process RNA molecules. In more
detail:
The nucleolus is the site of ribosome assembly, and forms in response to
transcription of ribosomal DNA (rDNA), the genes that encode ribosomal RNA. Preribosomal RNA (pre-rRNA) units associate with the small
nucleolar RNAs (snoRNAs), and are processed to form mature rRNAs.
Pretransfer RNAs (pre-tRNAs) are imported into the nucleolus and
processed there.The mature rRNAs associate with the tRNAs and a large
ensemble of ribosomal proteins to form ribosomes. Once assembled the
ribosomes are exported to the cytoplasm.
Cajal bodies (CBs) are the sites of assembly of transcription complexes,
and contain high concentrations of RNA Pol I, Pol II, and Pol III. Recall
that there are three kinds of eukaryotic polymerases. RNAP Pol II transcribes pre-mRNAs and most splicing RNAs. The other RNA polymerases transcribe rRNA subunits, pre-tRNAs, and the U6 splicing
RNAs. A cell may contain up to ten Cajal bodies. Once assembled within
a CB, pol I complexes translocate to the nucleoli, where they transcribe
rRNA genes. Pol III assemblages diffuse to the U6 small nuclear RNA
(snRNA) involved in splicing, 5S rRNA and tRNA transcription sites,
and Pol II complexes diffuse out of the CBs and over to mRNA and
snRNA transcription sites.
Interchromatin granule clusters (IGCs), or speckles, are distributed
throughout the inter-chromatin spaces. These organelles are enriched in
U1, U2, U4/U6, and U5 small nucleolar ribonuclear particles (snRNPs)
and other components of the splicing machinery. (These will be discussed
in detail in Chapter 15.) The localization of splicing factors within the
26
IGCs is not static, but instead changes in time in a way that reects the
transcriptional activity underway. Splicing factors diffuse in and out of
the granules to and from sites of transcription activity.
Figure 2.3. Amino acid structure: In amino acids, the bound organic groups,
denoted by the R symbols, are termed side chains. (a) Form of an uncharged
amino acid. (b) Form of a dipolar, or zwitterion, amino acid.
27
psi
phi
-57.8
-74.0
-57.1
-139.0
-47.0
-4.0
-69.7
+135.0
3.6
3.0
4.4
2.0
28
turns. Sequences that do not form one of these kinds of regular structures
are grouped together in a general category called random coils. The
arrangement of these features within the protein constitutes the proteins
secondary structure.
Alpha helices and beta sheets are stabilized by hydrogen bonds between
amide (NH) and carbonyl (CO) groups. In alpha helices, the hydrogen
bonds form between a carbonyl group on the ith residue and the amide
group on the (i + 4)th residue lying below it. In beta sheets, the hydrogen
bonds form between the carbonyl group lying on one strand and the amide
group situated immediately adjacent to it on the other strand. The beta
strands can be oriented in either a parallel or an antiparallel manner. Some
proteins are mostly alpha helix; other mostly beta sheet, and some are a
mixture of the two. These are referred to as a/b if the two types of structural element are mixed and a + b if they remain distinct.
Alpha helices are about 10 residues in length and these structures
account for about a third of the amino acid residues in a typical protein.
Beta sheets are typically 6 amino acid residues in length and they account
for a quarter of the residues.Turns allow the chain to reverse direction.They
along with loops are usually located on the protein surface and thus contain
polar and charged residues. Protein structures tend to be compact with little
space left open in the interior. Hydrogen bonds formed in the protein interior neutralize buried polar groups. Structural compactness and the use of
hydrogen bonds to neutralize interior polarity drive the formation of the
alpha helices and beta sheets.
Figure 2.5. Ramachandran plot: Shown is a stereotypic contour plot of the distribution of main chain torsion angles j and Y determined from high-resolution Xray crystallographic data. Darker shading denotes more highly favored torsion angle
combinations. Blank (white) regions represent conformations that are sterically
forbidden. The most favorable conformations for alpha helices and beta sheets are
concentrated into the three main regions. Beta sheet torsion angles appear as a
double-peaked distribution in the upper left quadrant of the plot. The distribution
of torsion angles for right handed alpha helices peaks in the lower left quadrant,
and that for left handed alpha helices peaks in the upper right quadrant. Each of
the 20 amino acids populates similar, but slightly different, portions of the
Ramachandran plot.
29
Figure 2.6. Topology of two common structural motifs: (a) The four-helix bundle,
and (b) the Greek key. In the plots, cylinders represent alpha helices, and arrows
oriented in the amino-to carboxyl-terminal direction denote beta strands. The
helices and strands are connected by short turns and longer loops. The four-helix
bundle can be regarded as a pair of helix-turn-helix structures. The Greek key is
assembled from two pairs of antiparallel beta strands.
30
tures pack into one of a small number of geometries and the chains assume
one of an equally small number of topologies. For example, beta sheets form
layered structures with either alpha helices or other beta sheets on their
faces. Alpha helices either distribute themselves about a core or form
layered structures. The packing of helices about a core can be described by
regular polyhedra. Three helices pack into an octahedron, four helices
describe a dodecahedron, ve helices form a hexadecahedron, and six
helices pack into a icosahedron. The packing of sheets is more variable.
Most beta sheets pack into two-layered structures, with the sheets either
aligned or orthogonal. Some beta sheets, notably in a/b proteins, form
barrels. In barrel patterns the b sheets coil around to form a cylinder and
the a helices are arranged on the cylinder surface.
The rules for chain topology state that knots in the polypeptide chain do
not form nor do secondary structure elements cross through one another.
Instead, the chain topology is minimally convoluted. Secondary structure
elements that are adjacent in the polypeptide sequence prefer to pack in
an antiparallel manner, and groupings of the form b-X-b, where X is either
an a-helix or a b-strand in an adjacent sheet, are right handed. The helices
pass through the folding domain and so the connections between helices
form on the outside along the ribs of the polyhedra. Beta sheet topology
forms hairpin, Greek key, or jellyrolls, depending on the number of strands
(respectively, two, four, six).
31
Figure 2.7. Domain organization of the Src protein, a nonreceptor tyrosine kinase:
It consists of four domainsan N-terminal SH3 domain, a C-terminal SH2 domain,
and two catalytic domains, connected by linker segments. Amino acid residues that
form well-dened secondary structure elements are drawn as (dark) coiled ribbons
and as (grey) planar ribbons or arrows. The coiled ribbons, like the cylinders used
in the previous gure, denote alpha helices. The planar ribbons or arrows form
layers that denote beta sheet secondary structures, either parallel or antiparallel.
The SH2 domain is of the form a + b; its secondary structure resembles a twolayered sandwich with two short alpha helices plus a central beta sheet. The SH3
domain contains a pair of antiparallel beta sheets. Tyrosine kinases catalyze the
transfer of phosphoryl groups to selected tyrosine residues. As can be seen in the
gure the catalytic domains are mostly alpha helical, except for the N-lobe, where
a prominent beta sheet component is present. Tyrosine kinases will be discussed in
Chapter 11. The gure was generated using Protein Explorer with the Brookhaven
Protein Data Bank (PDB) entry (accession number) 2Src containing the atomic
coordinates of Src determined by x-ray crystallography (to be discussed in
Chapter 3).
dened by the presence of one or more of the domains listed in the rst
column. The sizes vary from as few as 25 to 33 amino acid residues for the
minidomains to 120 amino acid residues for the PH domain. The domains
are not mutually exclusive, and a single protein will usually contain one or
more of several different kinds of domains. Minidomains are often arranged
in the proteins as tandemly repeated elements. Including the term repeat
as part of the name reects this propensity.
Several kinds of changes have taken place in the human genome. New
families of vertebrate genes appear that encode proteins belonging to the
immune and nervous systems, and there is a new family (KRAB) of zinc
nger transcription factors. A more widespread kind of change is the creation of new architectures, that is, of new arrangements of domains that can
subsume new functions. A third type of change is the large expansion of
existing families. Listed in Table 2.2 are representative examples of each of
32
Table 2.2. Commonly encountered domains in the human: Sizes of the domains
are given in terms of the numbers of amino acid residues. The fourth column lists
the number of genes encoding the domains. All of these domains have signaling
roles. The last column indicates the most prominent activities associated with proteins containing these domains. Im: immune system function; Extra: Extracellular
adhesion; Trans: transcription regulation.
Domain
Immunoglobulin
EGF-like
Fibronectin Type III
EF hand
Ankyrin repeat
Leucine-rich repeat
Cadherin
WD-40 repeat
Pleckstrin homology
Src homology 3
Src homology 2
PDZ
C2H2 zinc nger
Homeobox
RING nger
Krueppel-associated box
Symbol
Size
Genes*
Function
Ig
EGF
Fn3
EF
Ankyrin
LRR
Cadherin
WD40
PH
SH3
SH2
PDZ
C2H2
Homeobox
RING
KRAB
100
35
90
40
33
25
110
50
120
60
100
80
30
60
50
75
765
222
165
242
276
188
114
277
193
143
119
162
706
267
210
204
Im, Extra
Im, Extra
Im, Extra
Im, Extra
Im, Extra, Signal
Im, Extra, Signal
Im, Extra, Signal
Signal
Signal
Signal
Signal
Signal
Trans
Trans
Trans
Trans
* Data from International Human Genome Sequencing Consortium [2001]. Nature 409:
860921; Venter JC et al. [2001]. Science, 291: 13041351.
33
widely encountered in the immune system, and in these cases, unlike the
ion channels, the different chains may perform distinct functions.
Some of the large signaling proteins that reside in the cytosol and serve
as central organizers of the signal pathways are constructed from multiple
subunits. Specic functions are sequestered in the different subunits of the
proteins, and the ability of the proteins subunits to associate and dissociate from one another in a key part of how the signaling system works. Alternative splicing is often used to generate different forms of specic subunits.
This makes possible the creation of many different combinations of subunits; each one specialized for a specic cell or tissue type. By this means
many proteins of a similar kind, but each performing a slightly different
task, can be constructed from a small instruction set.
34
Farnesyl
Geranylgeranyl
Outer Leaet
Glycosylphosphatidylinositol
(GPI)
Attachment
35
Figure 2.8. Acyl anchors covalently attached to membrane proteins: (a) A 16-C
palmitoyl anchor is attached to a cysteine residue. (b) A 14-C myristoyl anchor is
covalently attached to a glycine residue.
Figure 2.9. Prenyl anchors covalently attached to membrane proteins: (a) A 15-C
farnesyl anchor is attached to a cysteine residue. (b) A 20-C geranylgeranyl anchor
is covalently attached to a cysteine residue.
36
37
this way, a single protein performs multiple tasks within a single signaling
pathway. Because several signaling intermediates are eliminated, the signaling process is a rapid one requiring few steps to go from the initiating
sensing stage to a terminating control point.
Proteolytic processing is not restricted to membrane proteins. It is utilized heavily to change the properties of cytosolic proteins from immobilized forms to mobile ones. Proteolytic processing is a key component of
signaling pathways involved in regulating the cell cycle, embryonic development, and immune function. The mechanism is similar in all of the
pathways. A crucial signaling element is parked in a specic location in the
cell through its binding to an inhibitory protein. In response to activating
signals, proteolytic enzymes (the 26S proteosome) chop up the inhibitory
proteins, thereby freeing the signaling proteins for movement into the
nucleus where they promote gene transcription. These mechanisms will be
discussed in more detail when the specic pathways in which they appear
are examined.
38
Figure 2.11. Covalent attachment of methyl and acetyl groups to arginine and
lysine side chains: Amino groups located at the ends of arginine and lysine side
chains provide sites for attachment of one or more methyl or acetyl groups.
39
nitrogens are present at the ends of arginine side chains, allowing for a
symmetric distribution of methyl groups between the two nitrogens, or,
alternatively, for one or the other of the nitrogens to have most if not all of
the methyl groups.
The enzymes that catalyze the transfer of methyl groups to arginine
and lysine residues are referred to as arginine methyltransferases and as
lysine methyltransferases, respectively. These enzymes use an endogenous
cellular molecule called S-adenosyl-l-methionine, or SAM, as the methyl
group donor. SAM is synthesized in cells of the body from methionine
and ATP, and readily donates its methyl group to the guanidine groups
on the arginine side chains and to the amino groups on the lysine side
chains.
The amino groups at the end of lysine side chains are the main
attachment sites for acetyl groups. The enzymes responsible for catalyzing
the transfer of acetyl groups to lysines are known as acetyltransferases.
These enzymes use acetyl coenzyme A (acetyl CoA) as the acetyl group
donor. The acetyl group is attached by a sulfur atom to CoA forming a
high-energy thioster bond, making it easy to transfer to acceptors such as
lysine.
40
41
Figure 2.13. Covalent modications of histone tails: Shown are regulatory sites
modied by acetylation (A), methylation (M), phosphorylation, or ubiquitination
(U). As is customary the sites are numbered in ascending order starting from the
N-terminal. One-letter codes for the amino acids are arginine (R), lysine (K), and
serine (S). (Three- and one-letter abbreviations for the amino acids are listed in
Table 4.1.)
lessened allowing for a greater access of the DNA to transcription regulators. Similarly, addition of net negative charge through phosphorylation on
serine residues also serves to decondense chromatin.
Many of the proteins that act in a supporting role to help activate or
suppress transcription interact directly with the histones rather than with the
DNA. Some of these enzymes function as histone acetyltransferases (HATs)
or alternatively as histone deacetylases (HDACs), adding or removing
acetyl groups from histone tails, using acetyl coenzyme A (acetyl-CoA) as
the donor. Other coregulatory enzymes operate in a similar manner, adding
or removing methyl groups or phosphoryl groups or SUMO groups or ubiquitin groups.
42
General References
Alberts B, Johnson A, Lewis J, Raff M, Roberts K, and Walker P [2002]. Molecular
Biology of the Cell, 4th edition. New York: Garland Publishers.
Matthews CK, van Holde KE, and Ahem KG [2000]. Biochemistry, 3rd edition. San
Francisco: Pearson Benjamin Cummings.
Stryer L [1995]. Biochemistry, 4th edition. New York: W.H. Freeman and Company.
Nuclear Organization
Lamond AI, and Earnshaw WC [1998]. Structure and function in the nucleus.
Science, 280: 547553.
Lewis JD, and Tollervey D [2000]. Like attracts like: Getting RNA processing
together in the nucleus. Science, 288: 13851389.
Chromosome Organization
Cremer T, and Cremer C [2001]. Chromosome territories, nuclear architecture and
gene regulation in mammalian cells. Nat. Rev. Genet., 2: 292301.
Manuelidis L [1990]. A view of interphase chromosomes. Science, 250: 15331540.
Protein Organization
Hovmller S, Zhou T, and Ohlson T [2002]. Conformations of amino acids in proteins. Acta Cryst., D58: 768776.
Kleywegt GJ, and Jones TA [1996]. Phi/Psi-chology: Ramachandran revisited. Structure, 4: 13951400.
Post-Translational Modications
Fortini ME [2000]. Fringe benets to carbohydrates. Nature, 406: 357358, and references cited therein.
McLaughlin S, and Aderem A [1995]. The myristoyl-electrostatic switch: A modulator of reversible protein-membrane interactions. Trends Biochem. Sci., 20:
272276.
Milligan G, Parenti M, and Magee AI [1995]. The dynamical role of palmitoylation
in signal transduction. Trends Biochem. Sci., 20: 181186.
Peschon JJ, et al. [1998]. An essential role for ectodomain shedding in mammalian
development. Science, 282: 12811284.
Resh MD [1999]. Fatty acylation of proteins: New insights into membrane targeting of myristoylated and palmitoylated proteins. Biochim. Biophys. Acta, 1451:
116.
Problems
43
Regulated Proteolysis
Brown MS, et al. [2000]. Regulated intramembrane proteolysis: A control mechanism conserved from bacteria to humans. Cell, 100: 391398.
Maniatis T [1999]. A ubiquitin ligase complex essential for the NF-B, Wnt/Wingless,
and Hedgehog signaling pathways. Genes Dev., 13: 505510.
Townsley FM, and Ruderman JV [1998]. Proteolytic ratchets that control progression through mitosis. Trends Cell Biol., 8: 238244.
Sumoylation
Mller S, et al. [2001]. SUMO, ubiquitins mysterious cousin. Nature Rev. Mol. Cell
Biol., 2: 202210.
Histone Modications
Grunstein M [1997]. Histone acetylation in chromatin structure and transcription.
Nature, 389: 349352.
Jenuwein T, and Allis CD [2001]. Translating the histone code. Science, 293: 1074
1080.
Problems
These problems require use of a PC to run the Protein Explorer, the free
software used to model the Src protein as shown in Figure 2.7. The rst step
is to install Chime to enable your browser to talk to the Protein Explorer.
Then bring up the Protein Explorer. (You may want to save the URL under
your Favorites pull-down.)
2.1 Bring up the atomic coordinates for the Src protein displayed in Figure
2.7. Its Protein Data Bank (PDB) accession number is 2src. Next
remove the water molecules and ligands leaving just Src. Then display
the image in the form shown in Figure 2.7. Hint: Go to Quick Views
and use the Display feature to bring up the cartoon depiction of
the secondary structure. Then drag on the image to rotate into the orientation presented in Figure 2.7.
2.2 Click on Mol Info and then the Header button to bring up additional information about the protein Src. Note the list of amino acids
comprising the primary structure.
2.3 Returning to the image of the Src protein. Bring up a backbone
display of the protein. What does this image show?
2.4 Now, bring up a ball-and-stick model of Src. What do the balls represent and what do the sticks denote? Click on some of the balls shown
in the image. What happens when this is done?
2.5 Change the display to a space-ll model. What do the spheres now
mean; that is, what do their radii represent? How is hydrogen treated?
3
Exploring Protein Structure
and Function
46
Level of detail
X-ray crystallography
Circular dichroism
Atomic
Molecular
Intermolecular
Molecular
NMR spectroscopy
Atomic
Process
X-ray diffraction
Absorption of polarized
UV light
Visible light absorption
and emission
Absorption (IR) and
scattering (Raman) of IR
light
Nuclear spin ips
Table 3.2. Physical methods used to explore protein structure and interactions.
Experimental method
Yeast two-hybrid
Gel electrophoresis
Mass spectrograph
DNA microarrays
Level of detail
Intermolecular
Molecular, Intermolecular
Molecular
Cellular
Process
Protein-protein interactions
Mass/charge separation
Mass/charge separation
Complementary base-pairing
c
,
l
(3.1)
47
(3.2)
-27
Figure 3.1. Electromagnetic spectrum: Shown in the upper portion of the gure are
the types of radiation emitted. The kinds of transitions, or motions, that absorb and
emit the radiation are shown in the lower part of the gure. Vertical dashed lines
delineate the boundaries between the different regimes. The actual boundaries
between radio waves and microwaves, and between ultraviolet and X-rays, are not
sharp but instead the regimes merge into one another.
48
Molecules are not static entities but instead are in constant thermal
motion, gaining and losing energy through random collisions with other
molecules. The kinetic energy absorbed in the collisions is converted into
vibrations and rotations about bond angles. These so-called collective modes
can also be excited when the atoms and molecules absorb radiation in the
appropriate wavelength range. The correspondence between wavelength
regimes of electromagnetic radiation and protein motions (transitions) that
produce the radiation is presented in the lower portion of Figure 3.1.
Recall that electrons move in specic atomic and molecular orbits, each
with a well-dened energy. The lowest energy state of the electrons in their
various orbits around a nucleus is called the ground state, and all others are
referred to as excited states. Besides inducing vibrational and rotational
activity, electrons can transition into excited states when radiation of the
correct wavelength is absorbed. Electromagnetic radiation of well-dened
energies is involved whenever electrons, atoms, and molecules undergo
transitions from one state to another, either higher or lower. The energy of
the absorbed and emitted radiation is equal to the difference in energies
between the two energy levels (states), and its frequency (wavelength) is
given by the following form of Plancks formula:
E2 - E1 = DE = hn.
(3.3)
49
50
Figure 3.3. Schematic depiction of uorescence: Shown are sets of related states,
or bands, along with vertical arrows denoting transitions between states. From left
to right, a packet of electromagnetic radiation is absorbed by a uorophore, producing a transition from the ground state to an excited state. Shortly thereafter, the
uorophore deexcites back to the ground state, emitting a photon with a slightly
lower energy and longer wavelength. Energy not carried off by the emitted photon
is lost through radiationless transitions between vibrational states within the excited
state band and within the ground state band.
51
W.H. Bragg and his son W.L. Bragg formulated the basic theory of relating diffraction patterns to atomic positions in a crystal in 19121913. They
noted that a three-dimensional crystal could be viewed as a set of equidistant parallel planes. The conditions for maximal constructive interference
are twofold. First, the scattering from each plane must be a specular, or
mirror, reection in which the angle of incidence equals the angle of reection. This situation is depicted in Figure 3.4. Second, the X-ray wavelength
l, distance between parallel planes d, and angle of reection q, obey the
relationship known as Braggs law:
2d sin q = nl.
(3.4)
In Eq. (3.4), n is an integer that can take on the values 1, 2, 3, and so on.
When n = 1, the spots of light are known as rst order reections, and when
n = 2, they are called second order reections. First order reections are
more intense than second order reections, and similarly for third and
higher order contributions.
The light spots are produced when the light waves arrive at the detector
in phase with one another and thus constructively interfere. In an X-ray
diffraction experiment, the intensities and positions of the light spots are
recorded. The diffraction patterns are converted into electron density maps
through application of a mathematical operation known as a Fourier transform. Several tens of thousands of reections are collected in a typical
X-ray diffraction experiment. Computer programs, taking as input the
resulting electron density map and knowledge of the primary sequence, are
52
Figure 3.5. X-ray crystallography: A unit cell is shown containing the molecule to
be studied. It is replicated many times in the crystal sample. X-rays scattered from
the atoms in the crystal produce a complex three-dimensional pattern of light and
dark spots in the detector called reections. The patterns of light and dark spots are
a consequence of the way the waves from different atoms in different locations in
the unit cell interfere with one another. A typical diffraction pattern produced in
one-dimension from a pair of atoms is depicted in the gure. As can be seen, there
is a series of peaks where the waves constructively interfere, and each peak is separated from the next by a trough where the waves from the two atoms destructively
interfere. These reections are then converted into an electron density map. A
typical portion of an electron density map is depicted. The map, a contour plot,
shows peaks where the electron density is high and valleys or open areas where the
electron density is low. In the model-building portion of the data analysis, the threedimensional structure of the molecule is deduced using computer programs.
53
54
Figure 3.6. Splitting of energy levels of a spin 1/2 particle by an external magnetic
eld: (a) In the absence of a magnetic eld, the energies of the spin up and spin
down states of the spin 1/2 particle are the same. The energy levels are no longer the
same in the presence of a magnetic eld. The energy of the particle whose spin is
aligned parallel to the magnetic eld, the spin +1/2 particle, is lower than the particle whose spin is aligned against the external magnetic eld, that is, the spin -1/2
particle. The energy levels are said to be split by the magnetic eld. (b) If the spin
1/2 particles are exposed to a source of electromagnetic energy, while in the magnetic eld, there will be a sharp absorption peak when the frequency of the electromagnetic wave exactly matches the frequency of the photon needed to trigger a
transition from the lower to the higher energy level.
55
Figure 3.7. Chemical shifts of hydrogen atoms in a CH3CH2 molecular environment: The arrangement of atoms in the two groups is depicted in the insert. The
chemical shifts are plotted with respect to a reference shift, taken as the zero of the
axis, and are given in units of parts per million (ppm).
56
of the nearby CH3 group induces the splitting of the single peak for CH2
into four peaks. Variation in peak heights is not arbitrary, but instead is a
direct consequence of the relative contribution to each peak from different
spin-spin interactions (see Problem 3.4).
eh
,
2 Mp
(3.5)
2m p B0
.
h
(3.6)
Nuclear magnetic resonance of protons in bulk matter was rst demonstrated in 1946 by Felix Bloch and, independently, by Edward M. Purcell.
57
Their efforts followed earlier work by Isidor I. Rabi, who discovered the
NMR effect in molecular beam experiments in 1937. Norman F. Ramsey,
working in Rabis laboratory, acquired the rst radiofrequency (RF) spectra
the following year, and developed the chemical shift theory in 1949. These
initial studies have evolved into a powerful technique used to study biomolecules in solution, and to a diagnostic tool, magnetic resonance imaging
(MRI), used in medicine.
58
Color
lex (nm)
lem (nm)
eBFP
eCFP
eGFP
eYFP
dsRed
blue
cyan
green
yellow
red
380
434
488
514
558
440
476
509
527
583
cell into which the vector was inserted) the resulting fusion protein will
contain the GFP covalently attached to the study protein, in a way that does
not interfere with the normal operation of that protein. The fusion protein
functions as a uorescent reporter. When excited either by a laser or by an
incandescent lamp, the protein will emit light and thus report its presence.
Fusion proteins made in this manner have been used to study how signaling proteins move about the living cell. They have been employed to visualize the subcellular compartmentalization of the signaling molecules, and
to view how the proteins move from one locale to another, in and out of
organelles, and back and forth to the plasma membrane.
In Aequorea victoria, GFP operates in close association with another
protein, aquorin, a naturally luminescent protein that emits blue light.
Energy is transferred in a radiationless manner from the uorophore of
aquorin to the uorophore of GFP. The GFP absorbs the blue light and in
turn emits green light. The process of radiationless energy transfer from one
uorophore to another is called uorescence resonance energy transfer
(FRET). This process can occur when the energy level differences involved
in emission from one uorophore overlap the energy level differences
involved in excitation of the second uorophore. That is, FRET can occur
if the donor emission spectrum overlaps the acceptor excitation spectrum.
59
the rst protein (the donor) to that of the second (the acceptor) when the
two proteins some into contact. These processes can then be studied with
light microscopy. The nature of the emitted light is sensitive to relative
orientation of the two uorophores and to their spatial separation. These
dependencies can be exploited in the design of the fusion proteins, so that
the emission spectra reect conformational changes and properties of the
protein-protein interactions.
The principle behind the use of FRET for studying protein-protein
interactions can be applied in a variety of ways to the study of signaling.
As was the case for protein-protein interactions, two particles are brought
into close proximity, one fused to a uorescent donor and the other to a
uorescent acceptor. The sought after signaling interactions are then
studied and quantied by measuring the changes in FRET efciency. When
the two uorophores are far apart the efciency is low, but this changes in
the presence of the interaction of interest. This is the basis for studying
post-translational modications such as proteolytic cleavage and phosphorylation; the movement of small signaling intermediaries such as cyclic
adenosine monophosphate (cAMP), Ca2+, and cytochrome c; and it has
been used to study the trafcking of proteins as they are secreted from the
cell.
Several examples illustrating how the FRET principle is applied are
presented in Figure 3.8. In the rst example, that of phosphorylation by a
protein kinase, a phosphorylation reporter is created. This (the reporter)
protein contains an amino acid sequence that is typically phosphorylated
by the kinase of interest. When the sequence is phosphorylated by the
kinase a second domain in the reporter protein recognizes and binds to the
phosphorylated sequence. The second domain contains a uorescent
protein that acts as the acceptor, while the rst domain contains the donor.
When the protein is phosphorylated by the kinase and the second domain
binds it, the uorescent acceptor is brought closer to the donor and the
FRET efciency increases.
A similar procedure is followed for proteins called cameleons, which
report the presence of calcium, an exceptionally important small signaling
molecule. In this case, calmodulin (CaM), a calcium-binding protein, is used
in constructing the reporter. Again a donor and acceptor are appended; the
donor is appended to CaM and the acceptor to a peptide known as M13
that binds tightly to CaM what the latter binds calcium. The third example
presented in Figure 3.8 represents the converse situation, that of proteolysis. To study proteolysis, a protein is created with donor and acceptor uorescent proteins. The two regions are connected to one another by an amino
acid sequence that is characteristically cleaved by the proteolytic enzyme
(protease) of interest. When the reporter protein is cleaved by the proteolytic protein, FRET ceases as the two uorescent proteins drift apart and
no longer interact.
60
Figure 3.8. Fluorescence resonance energy transfer (FRET) used to explore intracellular signaling: (a) PhosphorylationA laser emits radiation at 440 nm, triggering emission from a CFP at 480 nm in the left hand panel. The phosphorylation
recognition domain connected to the rst domain by a exible linker binds the phosphorylated protein following kinase activity in the right hand panel. These changes
result in FRET and an increased emission from the YFP at 535 nm. (b) Calcium
bindingIn the absence of calcium, a calcium-binding protein, calmodulin, is in a
conrmation that loosely tethers an M13 peptide. When it binds four calcium ions,
it assumes a dumbbell-like shape that induces a tight binding of the M13 peptide
resulting in FRET and increased emission at 535 nm. (c) ProteolysisIn the absence
of the protease, the two domains remain in close proximity and emit radiation at
535 nm. When the protease cleaves the linker, the two portions of the protein move
apart and no longer exhibit FRET.
61
the chirality condition. The left and right handed forms of chiral molecules
are collectively referred to as optical isomers or as enantiomers
nonsuperimposable mirror images of one another.
Chiral biological materials are sensitive to the chiral behavior of left and
right circularly polarized light and absorb the two light forms in different
ways. In circular dichroism (CD), differences in absorption between left and
right circularly polarized light are measured as a function of wavelength.
These measurements provide information on the secondary structure
content of the proteins.The measurements can be used to determine whether
the protein is mostly alpha helical, or whether it is made up mostly of antiparallel beta sheets and turns.Circular dichroism utilizes the ultraviolet (UV)
portion of the electromagnetic spectrum, typically in the range of 200 nm.
62
used since then, is the yeast two-hybrid system. This method involves the use
of designed transcription factors (the hybrids) and reporter genes. Reporter
genes are genes whose protein products have easily detected activities.Transcription factors are proteins that stimulate transcription. The transcription
factors used in the yeast two-hybrid approach activate the reporters.The rst
of these to be used is the Ga l4 protein in the yeast S. cerevisiae. This protein
stimulates transcription of the lacZ reporter gene that encodes the enzyme
b-galactosidase, whose activity can be easily measured (assayed).
Ga l4, like many transcription factors, contains a DNA-binding domain
and a transcription activation domain. (Transcription factors will be examined in detail in Chapter 16.) The modular organization of the Ga l4 protein
is exploited to create a pair of hybrid proteins. One hybrid consists of the
DNA-binding domain of Ga l4 fused to protein X. The other contains the
activation domain of Ga l4 fused to protein Y. The two hybrids are reintroduced into the yeast cell. If protein X (the bait) interacts with protein Y
(the prey) the Ga l4 DNA and activation domains will be brought into close
proximity and the pair will be able to stimulate transcription of the reporter
gene.
In the yeast two-hybrid method, different combinations of bait and prey
are tested to identify interacting proteins. A common approach to carrying
out large scale testing is to utilize two haploid yeast strains. One strain
contains the bait and the other the prey. The strains are then mated and the
interactions are determined by assaying the activity of the reporter gene in
the diploid strain.This variant of the two-hybrid method is called interaction
mating or mating assaying. It has been applied to several organisms whose
genomes have been sequenced. For example, several hundred proteinprotein interactions have been identied in this manner in S. cerevisiae.
63
64
(3.7)
The voltage V is usually kept constant; the radius of curvature r is determined by the geometric properties of the magnet, and the user varies B to
select different M/z ratios. In a further renement of the technique, a pair
of parallel electrodes can be added to the magnet. By adjusting the electric
and magnetic elds according to the expression r = 2V/E, the velocity
dependence can be removed so that all ionized proteins of a given M/z
value fall on the same point in the focal plane of the detector.
65
M z = 2V (t L) .
(3.8)
66
Circular Dichroism
Johnson WC [1990]. Protein secondary structure and circular dichroism: A practical guide. Proteins, 7: 205214.
Woody RW [1995]. Circular dichroism. Methods in Enzymology, 246: 3471.
2D Gel Electrophoresis
Gygi SP, et al. [1999]. Quantitative analysis of complex protein mixtures using
isotope-coded afnity tags. Nat. Biotechnol., 17: 994999.
Mass Spectrometry
Link AJ, et al. [1999]. Direct analysis of protein complexes using mass spectrometry, Nat. Biotechnol., 17: 676682.
Yates JR, 3rd [1998]. Mass spectrometry and the age of the proteome. J. Mass
Spectrom., 33: 119.
Problems
67
Problems
3.1 (a) The emission of laser light at 440 nm for use in FRET was discussed
in Section 3.10. What is the energy of these photons in (i) eV, and
(ii) kcal/mol? (b) What is the energy of a 1.5 X-ray in these two sets
of units? (c) What are the corresponding frequencies of the photons at
440 nm and 1.5 .
The following list of physical constants and conversion factors may be
helpful:
Physical constants and conversion factors
Avogardos number N = 6.022 1023/mol
Velocity of light c = 2.9979 1010 cm/s
Plancks constant h = 6.626 10-27 erg sec
1 eV = 1.6 10-12 erg
1 cal = 4.184 joules .004 4.184 107 ergs
3.2 Using the gure shown below, derive Braggs law, Eq. (3.4).
Start with the relationship that the increase in path length between the
two scattered rays shown in gure must be an integral multiple of the
wavelength, that is, the waves must be in phase with one another in
order to constructively interfere. In terms of the notation of the gure
this condition is
r + x = nl .
Hint: Make use of the trigonometric identity:
2 sin 2 q - 1 = cos (180 -2q) .
3.3 Show that the nuclear magneton
m N = 5.05 10 -27 J T .
68
In the unit J/T, J denotes Joules and the symbol T denotes Tesla, the SI
unit for B-elds. This quantity has dimensions of kilograms per second
per second per ampere; that is 1 T = 1 kg s-2 amp-1. Use values for physical constants from the table in Problem 3.1 and the one shown below.
(Note: The ampere is a unit of current; that is, 1 amp = 1 coulomb per
second).
Physical constants and conversion factors
Charge on an electron e = 1.6 10-19 coulombs
Mass of a proton Mp = 1.672 10-27 kg
3.4 Recall from Figure 3.7 that the CH2 group splits the single peak for the
hydrogen atoms in CH3 into three peaks, and the CH3 group splits the
single hydrogen peak for CH2 into four peaks. In the gure shown
below, the spin-spin effect of the CH2 group on the CH3 group is shown.
The two hydrogen atoms in CH2 can each have their spins either
aligned with the external eld (+) or have their spins aligned opposite
(-). When opposite they shield the magnetic eld and reduce the splitting, while aligned they add to the eld and increase the splitting. There
are three different spin-spin combinations: both negative, one positive
and one negative, and both positive. This gives rise to the three peaks.
Since there are two ways of arriving at a [+, -] combination and once
each for the others the peak intensities are in a 1 : 2 : 1 ratio as illustrated
in Figure 3.7.
(a) In this problem, write down the possible spin combinations for the
CH3 group that splits the peak for CH2 into four peaks shown in Figure
3.7, and give the ratios of the peak heights. (b) From the results for two
and three hydrogens, infer the splittings of a hydrogen peak produced
by four nearby hydrogen atoms. How would their peak heights vary?
Problems
69
3.5 Recall from the discussion on mass spectrometers that the beams of
charged particles are subjected to rst an accelerating voltage allowing
the particle to attain a kinetic energy equal to
zV = mu 2 2.
When the particles feel the magnetic eld the force zuB is exactly balanced by the centrifugal force:
zuB = mu 2 r .
(a) From these two relationships derive Eq. (3.7). (b) Then derive the
time-of-ight expression given by Eq. (3.8).
4
Macromolecular Forces
72
4. Macromolecular Forces
Table 4.1. Physical properties of amino acid residues:
Three- and one-letter abbreviations (codes) are listed in
column 2. Masses are given in column 3 in Daltons. The
volumes presented in column 4 are in cubic angstroms.
Amino acid
Code
Mass (Da)
Volume (3)
Alanine
Arginine
Asparagine
Aspartic acid
Cysteine
Glutamic acid
Glutamine
Glycine
Histidine
Isoleucine
Leucine
Lysine
Methionine
Phenylalanine
Proline
Serine
Threonine
Tryptophan
Tyrosine
Valine
Ala-A
Arg-R
Asn-N
Asp-D
Cys-C
Glu-E
Gln-Q
Gly-G
His-H
Ile-I
Leu-L
Lys-K
Met-M
Phe-F
Pro-P
Ser-S
The-T
Trp-W
Tyr-Y
Val-V
71.08
156.2
114.1
115.1
103.1
129.1
128.1
57.05
137.1
113.2
113.2
128.2
131.2
147.2
97.12
87.08
101.1
186.2
163.2
99.13
89.3
190.3
122.4
114.4
102.5
138.8
146.9
63.8
157.5
163.0
163.1
165.1
165.8
190.8
121.3
93.5
119.6
226.4
194.6
138.2
longest side chains belong to arginine and lysine, while the most massive
are arginine, tyrosine and tryptophan. The lengths of the side chains vary
by an order of magnitude and the masses of the amino acids, which are
listed in Table 4.1, differ by more than a factor of three.
The amino acids used to make proteins vary not only in size, but also in
shape. Some amino acids have open side chains (alipathic); others contain
closed rings (aromatic); and one amino acid, proline, has a side chain that
closes back onto the main chain nitrogen. That the amino acids vary in size
and shape is important for their packing. These variations make possible
the close packing of amino acids in the core of the folded protein. The
resulting interior packing densities approach those of organic solids.
73
Table 4.2. Electrostatics and bonding propensities of the amino acids: The polar
amino acids listed in column 3 are of three kinds. Some are basic (arginine, histidine,
and lysine); others are acidic (aspartic and glutamic acids); and the remainder are
uncharged polar molecules. Histidine is only weakly basic and is often considered
a nonpolar amino acid, along with proline.
Amino acid
Alanine
Arginine
Asparagine
Aspartic acid
Cysteine
Glutamic acid
Glutamine
Glycine
Histidine
Isoleucine
Leucine
Lysine
Methionine
Phenylalanine
Proline
Serine
Threonine
Tryptophan
Tyrosine
Valine
Nonpolar
Polar
H-bond
Salt Bridge
*
*
*
S Bridge
*
*
*
*
*
*
*
*
*
*
*
*
*
*
*
*
*
with other biomolecules, and how proteins interact with their aqueous
environments.
The results of grouping the 20 amino acids according to their charge and
bonding properties are presented in Table 4.2. Columns 2 and 3 in the table
group the amino acids according to whether they are polar or nonpolar. The
signicance of this partitioning is that amino acids with nonpolar side chains
are hydrophobic, and those with polar side chains are hydroplilic. The
distinction between the two kinds of interaction arises from the binding
preference of water for the amino acid. In the case of a hydrophilic (waterloving) amino acid, a water molecule would rather bind to the amino acid
than to another water molecule. In the case of a hydrophobic amino acid,
a water molecule prefers another water molecule to the amino acid.
Nonpolar groups tend to come together in water, not because of an
attraction for each other but rather because their clustering enables water
molecules to make the maximum number of contacts with each another.
The term hydrophobic interactions denotes the process whereby water
molecules come together so that small nonpolar molecules and nonpolar
portions of large molecules minimize their contacts with water. The
propensity for water molecules to come together is a reection of their
74
4. Macromolecular Forces
75
76
4. Macromolecular Forces
Table 4.3. Atomic radii: Listed are average values for
atomic radii deduced from X-ray crystallography data
for a variety of atomic groups in proteins. All atomic
radii are in angstroms (). The radii listed in columns
2 through 5 are for covalent, coulombic, hydrogenbonded and van der Waals interactions, respectively.
Atom
rcov
rcoul
rH
rvdW
Nitrogen
Oxygen
0.70
0.65
1.45
1.40
1.55
1.40
1.70
1.50
atomic radii and bond energies behave in a systematic way with respect to
one another. The stronger the bond the closer together the two atoms will
be. Covalent bonding strengths can be as high as 100 kcal/mol or more. This
is at least an order of magnitude greater than any of the noncovalent bond
forms. As a result the bond lengths are about a factor of two shorter than
those of any of the weaker bonding forms. The strongest of the noncovalent forces are the coulombic interactions. These can be as much as 5 kcal/
mol. The hydrogen bonds are somewhat weaker yet, broadly distributed in
the range of 2 kcal/mol. The weakest of the forces are the van der Waals
interactions. These are not more than about 1 kcal/mol, barely above typical
thermal energies of 0.6 kcal/mol.
The three kinds of interactions just discussed, hydrogen bonds, coulombic attractions and repulsions, and van der Waals forces, are all far weaker
than the covalent bonds that underlie the protein backbone. Rather than
forming a few strong covalent bonds proteins interact with one another by
forming multiple weak bonds that can be easily broken and reestablished
in the same or in different ways.
77
By analogy with the characterization of disfavored water-protein interactions as a hydrophobic effect, disfavored osmolyte-protein interactions
are called osmophobic interactions. This effect manifests itself as a burial of
the protein backbone to shield it from the osmolytes. The gain in stability
from avoiding contacts between the backbone atoms and the osmolytes
more than compensates for the attractive effects between the side chains
and the osmolytes. By increasing the concentration of osmolytes when
stressed, a cell supplies a driving force that promotes the folded state over
the unfolded one.
Function
Coordinates activities of adjacent domains within a protein
Enables subunits on pore- and channel-forming proteins to work as
a single unit
Coordinates activities of cytosolic protein subunits that transiently
associate
Communication between proteins
Communication between proteins and regulatory sequences in
DNA molecules
Communication between proteins and RNA molecules
Communication between proteins and membrane lipids
Recognition of cellular and ECM carbohydrates
78
4. Macromolecular Forces
The other categories of interfaces operate between proteins and the different kinds of cellular biomoleculesproteins, DNA, RNA, lipids, and carbohydrates. These interfaces make possible the binding of proteins to their
ligands, transcription factors to DNA, and splicing factors to RNA. They
mediate interactions with the lipid membrane bilayer and are responsible
for the coordinated activities of signal complexes. From the viewpoint of
signaling the most widely encountered and studied interfaces are the
protein-protein and protein-DNA interfaces. Carbohydrate interfaces are
widely encountered in cells of the immune system, and these will be discussed in Chapter 10, which is devoted to cell adhesion and motility. Lipid
interfaces are crucial for a variety of cellular processes, including signaling.
These interfaces will be examined in detail in Chapter 8, where membrane
lipid composition and lipid signaling are explored.
Recall from Chapter 2 that proteins involved in signaling are frequently
post-translationally modied. Some amino acid residues acquire phosphoryl
groups, while others gain acetyl groups or methyl groups. Some protein
interfaces are sufciently precise in their binding afnities that they can
distinguish whether these groups are present or absent. As a result, an
upstream signaling event, where a group is added, can be followed by a
downstream event, which involves the recognition of that binding site.
79
(4.1)
Several different kinds of motions of atoms about their bonds are possible.
As depicted in Figure 4.1, there are stretching motions, bending motions,
and torsions about the bond axis.
80
4. Macromolecular Forces
Figure 4.1. Motions about groups of two, three, and four covalently bonded atoms:
Displayed in the upper portion of the gure are bond-stretching motions of a pair
of covalently bonded atoms along their bond axis. Shown in the middle panel are
bending motions involving three bonded atoms forming an angle q. Displayed at
the bottom are groups of four atoms, where the leftmost atom has rotated about
the middle bond axis.
K (b - b ) + K (q - q
= + K (1 - cos (nf + d)).
U bonded =
bonds
eq
angles
torsions
eq
(4.2)
81
Figure 4.2. Bond stretching: Plotted are the potential energies associated with bond
stretching. The harmonic potential has a minimum at the equilibrium bond length.
It increases as the bonded atoms are pushed along the bond axis towards one
another, so the bond is compressed, and as they are pulled apart, so the bond is
stretched out. For the stretch spring constant, Kb = 400 kcal/(mol 2), and for the
equilibrium bond length, beq = 1.3 .
82
4. Macromolecular Forces
Figure 4.3. Bond bending: Plotted are the potential energies associated with rotations of the angle made by three atoms covalently bonded to one another. In a
manner analogous to bond stretching, there is an equilibrium bond angle where the
potential energy is at a minimum. As the rotation angles are either widened out
or squeezed in, the potential energy rapidly increases. The bending spring constant
Kq = 40 kcal/(mol rad2), and equilibrium angle qeq = 120 deg.
Figure 4.4. Bond torsions: Plotted are the periodic contribution to the potential
energy from torsion (dihedrals) rotations of covalently bonded sets of four atoms.
Calculations were done using a Kj with four paths so that 3.625 kcal/mol is the constant in front of the expression. Parameters values used were n = 2 and d = 180 deg.
involving the twelfth power of the radius is a repulsive one. As can be seen
in the gure there are two distances of interest. One of these is the equilibrium radius r0, the location where the potential has its minimum and
where the forcethe derivative of the potentialis zero. The other key dis-
83
Figure 4.5. Van der Waals interactions: Plotted are the van der Waals potential
energies in kcal/mol using a well depth e = 0.19 kcal/mol and a radius r0 = 3.59 ,
corresponding to nitrogen and oxygen contact radii of 1.7 + 1.5 = 3.2 .
tance is the radius at which the repulsion exactly cancels out the attraction
so that the net potential is zero. This repulsion is generated by the interpenetrating electron clouds. This contact distance is the van der Waals
radius. As can be seen in the plot, any further interpenetration is strongly
resisted by the rapidly increasing repulsive force arising from the Pauli
exclusion.
6
r0 12
r0 qi q j
+
U nonbonded = e
-2
r 4pe 0 r
i j r
(4.3)
84
4. Macromolecular Forces
integration. The forces F are the spatial derivatives of the potential energies U presented in Eqs. (4.2) and (4.3). These quantities are then converted
to accelerations using Newtons second law, F = ma, where m is mass and a
is acceleration. Once this is done the equations of motion are solved to give
a picture of how the system evolves over time. Numerical methods are used
to convert the equations of motion into a form suitable for numerical integration on a computer. A number of computer programs are in use that
enable the researcher to carry out MD simulations.
The last entry in the list of methods is quantum mechanics (QM). The
covalent bonds are only handled in an approximate way in the MM formalism. In a quantum mechanical approach, the empirical treatment of the
covalent bonds through the use of a Hookes law is replaced by an exact
quantum mechanical treatment. The quantum mechanical method is exact
and rigorous but is difcult to use in practice due to computational limitations. As computer technologies have advanced, quantum mechanical
studies have become more numerous. An often-used approach is to
combine the MM and QM formalisms, using one or the other of the two
methods for different aspects of the system under study.
85
works between water and DNA is essential for DNA stability. DNA denatures as it dehydrates. Hydrogen bonds between water and ssRNA are even
more numerous than in the case of dsDNA because of the single strand
character of the RNA leaves bases unpaired and there are additional ribose
oxygen atoms available for bonding.
General Reference
Brandon C, and Tooze J [1999]. Introduction to Protein Structure, 2nd edition. New
York: Garland Science Publishing.
86
4. Macromolecular Forces
Sheinerman FB, and Honig B [2002]. On the role of electrostatic interactions in the
design of protein-protein interfaces. J. Mol. Biol., 318: 161177.
Tsai J, et al. [1999]. The packing density in proteins: Standard radii and volumes.
J. Mol. Biol., 290: 253266.
Hot Spots
Bogan AA, and Thorn KS [1998]. Anatomy of hot spots in protein interfaces. J. Mol.
Biol., 280: 19.
Hu ZJ, et al. [2000]. Conservation of polar residues as hot spots at protein interfaces. Proteins, 39: 331342.
Problems
4.1 Atomic motions. Atoms in a protein are constantly in thermal motion.
Assuming an average energy kT of 0.6 kcal/mol for each atom, how fast
is a hydrogen atom moving? How fast is a carbon atom moving? How
long will it take for each of these atoms to move 1 , roughly one bond
length?
Problems
87
1
1 d
Fi =
Ui .
mi
mi dri
r (t + Dt ) = 2r (t ) - r (t - Dt ) + a(t )(Dt ) .
Note that the velocities do not appear in this expression. The positions
at time t + Dt are computed from the positions at the present (t + Dt)
and previous (t) times and from the accelerations at the previous (t)
time. Some algorithms use the velocities explicitly in the computations.
One of these is the velocity Verlet algorithm. Its form is
v(t + Dt ) = v(t ) + (1 2)[a(t ) + a(t + Dt )]Dt .
By making use of the appropriate expansions, and combining terms,
derive both of these Verlet algorithms. What might be an appropriate
time step size? (Hint: Think about the results from Problem 4.1.)
5
Protein Folding and Binding
The world contains a myriad of biological systems. All exhibit a considerable degree of order.They are organized in a hierarchical manner, and order
is present at all levels of the hierarchy. From hydrogen, carbon, nitrogen,
and oxygen, simple atomic groups are formed such as methyl (CH3) and
hydroxyl (OH). These groups are then used to form the basic building
blocks of cellssugars, fatty acids, nucleotides, and amino acids. Simple
sugars (monosaccharides) are organized into short chains (oligosaccharides) or longer ones (polysaccharides). Fatty acids form complexes such as
triglycerides and phospholipids. Nucleotides are used to make RNA and
DNA, and the amino acids give rise to polypeptides, or proteins.
Biological order does not arise out of some mysterious vital force, but
rather is a consequence of the laws of thermodynamics and the character
of the forces in our universe, their strength and their dependence on distance. At rst glance the emergence of highly organized biological entities
seems at odds with the second law of thermodynamics. This law establishes
a thermodynamic arrow of timethe total disorder in the universe
increases as the universe ages, until a terminal stage of disorder is reached
in which the universe suffers a heat or entropy death. Yet, this rst impression is wrong: Order comes about not in spite of the laws of physics but
rather because of them.
Biological systems are open, continually exchanging matter and energy
with their surroundings.They generate order by taking in energy and releasing heat to their surroundings. They absorb radiant energy from sunlight
and from geothermal sources, and they take in foodstuffs that store energy
in high energy chemical bonds. According to the second law of thermodynamics the amount of entropy, or disorder, in a cell and its surroundings
must increase during any process. Thus, the production of order within a
cell is accompanied by the creation of a greater amount of disorder outside
a cell. This is accomplished through the release of heat from the cell at the
same time that the order is produced. Biological entities are not only highly
ordered, but actively generate these states in order to survive and
propagate.
89
90
(5.1)
Energy will be gained by a system whenever energy ows into the system
due to temperature gradients and whenever the surroundings do work on
91
the system. Energy will be lost from a system whenever the system does
work on its surroundings and whenever energy is lost to the surroundings
due to thermal gradients. If we consider pressure-volume work then we may
write this as
dE = Q - PdV ,
(5.2)
92
Figure 5.1. Mass and thermal equilibration: (a) Mass equilibrationIn the left
panel, most of the mass is concentrated in the left compartment. The particles are
free to diffuse to and fro, and through the opening into the adjoining compartment.
Over time, the system will mass equilibrate. In the right panel, roughly half of the
particles are in each compartment. (b) Thermal equilibrationArrows denote
velocity; particles with longer arrows are moving with a greater velocity than those
with shorter arrows. In the left panel, all of the fast particles are in the left compartment, and all the slow particles are in the right compartment. As a result, the
temperature in the left compartment is higher than that in the right compartment.
Again, there is an opening, and the particles can freely move about. Over time, the
system equilibrates so that the temperature in each chamber becomes the same. In
the right panel, each compartment contains a similar mix of slow and fast moving
particles.
other. In the case of thermal equilibration, the temperatures in the two compartments initially differ. The compartment with the fast movers is at a
higher temperature than that containing the slow movers, but over time the
temperatures in the two compartments become the same.
93
(5.3)
If a living cell is to increase its internal order it must be in contact with its
surroundings to allow for the exchange of energy. If it is isolated from its
surroundings the amount of disorder within the cell can only increase since
the second part of the second law asserts that
dScell > 0.
(5.4)
(5.5)
It is now possible for dScell to become negative so that the amount of order
in the cell can increase. This can occur if the increase in disorder in the surroundings is greater than the production of order within the cell.
(5.6)
In this expression a minus sign has been introduced since Q represents the
amount of heat absorbed from the surroundings. Since at constant temperature and pressure Q = dH, the entropy of the surroundings is simply
dSsurr = -dH/T, and
94
(5.7)
(5.8)
Since the right hand side of this last equation is simply the negative of the
right hand side of the previous equation, combining the two expressions
yields the relation
TdStotal = -dG.
(5.9)
This is a remarkable result. It states that the net change in entropy can be
determined from an examination of the change in the Gibbs free energy of
the system alone. Thus, there is no need to evaluate the change in entropy
in the surroundings in order to determine whether a process will occur
spontaneously or not. For a process to occur spontaneously there must be
a net increase in the entropy, or equivalently, the Gibbs free energy of the
system must decrease:
dG < 0.
(5.10)
By considering the Gibbs free energy one can determine whether a process
will occur spontaneously or not. There are two parts to consideran energetic (enthalpic) piece and an entropic one. Crystalline solids are more
ordered than liquids, and liquids, in turn, are more ordered than the gaseous
phase of a given substance. A crystalline solid such as ice can spontaneously
melt to become liquid decreasing the system order. In this process the
energy of the system increases, but this increase is more than offset by the
accompanying increase in entropy or disorder. Conversely, a process that
increases the order within a system can occur spontaneously if the decrease
in entropy is compensated for by a decrease in internal energy. The most
favorable reactions are those where energetic and entropic changes are
aligned, and spontaneous processes do not occur at all when entropic and
energy changes both increase the Gibbs free energy.
95
sheets. The set of similar states into which protein folds is collectively
referred to as the native state. Conversely, the collection of states that a
newly synthesized protein, or an unfolded protein, populates is called the
denatured state. In a series of pioneering experiments in the 1950s and 1960s
Ansnsen showed that protein folding is a reversible process. By varying
conditions in the aqueous environment, proteins were made to go back and
forth between folded and unfolded congurations. Two main conclusions
can be drawn from his experiments. First, all the information needed for
folding is contained in the primary sequence. Second, the native and denatured states are thermodynamically stable statesthey are states of minimum Gibbs free energy.
The most important environmental or physical parameter inuencing
protein stability is temperature. (Two others are pH and salt concentration.)
The native state is a minimum in the Gibbs free energy at physiological
temperatures (and conditions). However, if the temperatures are elevated
above a critical temperature, the denatured state of a protein will lie at a
lower Gibbs free energy than the native state. The reason for this can be
discerned in an examination of the behavior of the two terms in Eq. (5.8)
for the Gibbs free energy. As the temperature is raised, the entropic contribution gains in importance relative to the enthalpic term. The entropic
term is a measure of the number of possible congurations for the main
and side chains. The entropy favors the denatured state because there are
many more ways for the side chains to arrange themselves when unfolded
than when tightly folded into a globular form. The enthalpic contribution,
on the other hand, strongly favors the native state. At low temperatures the
entropic contribution is still appreciable, but the enthalpic term predominates in this regime.
The property of stability is an important one. To be useful a state must
be stable long enough for the biological entity, whether it be a protein, a
DNA molecule, or some larger structure, to carry out its biological function
Such states must be stable in a thermodynamic sense. These states, once
formed, do not change appreciably in time. The effects of small perturbations and of thermal uctuations are rapidly damped out and the behavior
of the system is not appreciably altered. These are equilibrium states in the
language of thermodynamics.
While there are a multitude of states corresponding to the denatured
state, there are usually only a few similar states corresponding to the native
state and its conformation is essentially unique. This aspect is noted in Table
5.1. In order for the native state to be stable there must be an appreciable
gap in energy between the native state and nearby nonnative ones. When
the differences are appreciable, it is difcult for small perturbations and
thermal uctuations to induce transitions to the nearby higher energy
states. Whenever the energy gaps are small, the proteins will be only marginally stable.
96
Fast folding
Folding funnel
Kinetic trap
Native state
97
Figure 5.2. Energy landscapes: Each point in the cutaway views of the energy landscapes represents a possible conguration of the protein. In these 3D depictions, the
x, y-axes represent generalized coordinates indicative of the states of the protein.
The vertical axis denotes the potential energy, or equivalently, the enthalpy. The
overall shapebroad at the top and narrow at the bottomgives rise to the description of these surfaces as folding funnels. The funnel on the left is smooth and the
protein trajectories are straight, running down the funnel from the unfolded state
to the native state. The funnel on the right is slightly more complex. A ridge is
present which has to be surmounted before the protein can slide down to the native
state. The trajectories are longer and convolute slightly as they pass over the ridge
and then down the funnel. Even more complex funnels are possible, especially for
multidomain signaling proteins, in which there are mountains and valleys that have
to be traversed during folding.
landscape at a high potential energy and ends at the native state located at
the bottom of the landscape at a low potential energy.
The amount of time required for a protein to fold into its native state is
an important aspect of the process. This is referred to as a kinetic requirement. Not only must a protein fold into its native state, but also it must do
so in a physiologically reasonable time interval. The speed depends critically on the topography of the potential energy surface. If the surface is
studded with deep minima, and the folding trajectories pass close to them,
the rate of folding will be slow. In these situations, the protein will fall into
the minima and must escape before proceeding with its evolution towards
its native state. The deep minima are called kinetic traps because of their
slowing effects on the kinetics, or rates, of folding. Large and complex proteins, especially those involved in signaling pathways, tend to have rugged
landscapes containing minima surrounded by high barriers. On the other
hand, small single domain proteins often have landscapes that are fairly
smooth and these proteins fold rapidly. The difference between smooth and
rough funnels is highlighted in Figure 5.3.
98
Figure 5.3. Smooth and rough folding funnels: (a) Smooth funnel in which there
are few barriers and all of these are smaller in height than the thermal energy.
(b) Rough funnel possessing several barriers that are difcult to surmount and serve
as kinetic traps because their heights are much larger than the thermal energy.
One of the features present in the rough funnel depicted in Figure 5.3 is
that of a low-lying metastable state. Recall from the discussion at the end
of Section 5.5 that one of the conditions for stability of the native state is
that there be an appreciable energy gap between the native state and those
lying above it. This condition is violated in the rough landscape depicted in
Figure 5.3. The protein will spend an appreciable time in the nearby excited
state. Not only is that state not separated by a large energy gap, but also,
once in that state, the protein must surmount a kinetic trap to get back out
to the native state.
99
and cannot insert in the membrane. The consequence is that the proper
movement of chloride ions is impaired. Among the most prominent symptoms of the disease are the production of salty secretions by the sweat
glands and thick mucus secretions by the lungs.
Another example is how point mutations can lead to impaired folding
and disease is rhodopsin. This protein is found in rods in the retina, where
it functions as the phototransducer. When this protein is mutated in regions
away from the C-terminus it fails to fold properly and doesnt transduce
light. In retinitis pigmentosa, the name for the resulting disorder, the rods
die off; sight worsens progressively, and eventually the subject becomes
blind.
One of the most striking examples of how even a single mutation can
have disastrous consequences on protein folding is that of the blood oxygen
carrier, hemoglobin. In this protein, a substitution of valine for glutamic acid
in the beta chains leads to improper assembly of the four subunits. The
result is sickle cell anemia. The hemoglobin molecules are misshapen and
not only dont transport oxygen adequately, but tend to clump together
causing further impairments. The clumping of misfolded proteins is not
limited to hemoglobin. It is observed in a variety of neurological disorders,
as well.
100
Amyloid-forming protein
Amyloid b protein
Prion
Huntingtin
CuZn superoxide dismutase
Alpha synuclein
101
102
Distribution
GroEL in bacteria; Hsp60 in
mitochondria and chloroplasts
Hsp10
Group II Chaperonins
TriC/CCT chaperonins in
archaea and eukaryotic cytosol
Hsp70 Chaperones
Hsp70
Hsp40
Hsp90 Chaperones
Function
Protected environment for
folding, refolding, recovery
from stress
Co-chaperonin for Hsp60;
assists in folding substrates
bound to Hsp60
Protected environment for
folding, refolding, recovery
from stress
Regulates heat shock response,
stabilizes nascent chains, and
prevents aggregation
Co-chaperone for Hsp70;
regulates activity of Hsp70
Maturation of signal
transduction pathways
103
Figure 5.4. A GroEL-GroES Annsen cage for protein folding: Fourteen subunits
are arranged into two rinds of seven units each. Each chain contains equatorial,
intermediate, and apical domains. Polypeptide chains are inserted into the cage,
undergo a round of mechanical manipulations (i.e., the chains are annealed to
remove unfavorable bondings), and are ejected. Several rounds of these operations
may take place.
104
Figure 5.5. Low-lying ensemble of native and nearby state: These states determine
the binding properties of the proteins. Different ligands acting as environmental
factors select one or more of these preexisting states when they bind the protein.
105
Motions of Proteins
Cavanagh J, and Akke M [2000]. May the driving force be with youWhatever it
is. Nature Struct. Biol., 7: 1113.
Feher VA, and Cavanagh J [1999]. Millisecond-timescale motions contribute to
the function of the bacterial response regulator protein Spo0F. Nature, 400:
289293.
Forman-Kay JD [1999]. The dynamics in the thermodynamics of binding. Nature
Struct. Biol., 6: 10861087.
Frauenfelder H, Sligar SG, and Wolynes PG [1991]. The energy landscapes and
motions of proteins. Science, 254: 15981603.
Kay LE, et al. [1996]. Correlation between dynamics and high afnity binding in an
SH2 domain interaction. Biochem., 35: 361368.
106
Binding Mechanisms
DeLano WL, et al. [2000]. Convergent solutions to binding at a protein-protein
interface. Science, 287: 12791283.
Freire E [1999]. The propagation of binding interactions to remote sites in proteins:
Analysis of the binding of the monoclonal antibody D1.3 to lysozyme. Proc. Natl.
Acad. Sci. USA, 96: 1011810122.
Hilser VJ, et al. [1998]. The structural distribution of cooperative interactions in
proteins: Analysis of the native state ensemble. Proc. Natl. Acad. Sci. USA, 95:
99039908.
Problems
107
Kumar S, et al. [2000]. Folding and binding cassettes: Dynamic landscapes and population shifts. Protein Sci., 9: 1019.
Ma B, et al. [2002]. Multiple diverse ligands binding at a single protein site: A matter
of pre-existing populations. Protein Sci., 11: 184197.
Teague S [2003]. Implications of protein exibility for drug design. Nature Rev. Drug
Dis., 2: 527541.
Problems
5.1 Entropy, density of states, and probabilities. Consider a system of N
atoms, each of which is placed in one of M bins. The atoms are labeled
by the bins into which they are placed, but are otherwise indistinguishable from one another. That is, n1 atoms are placed in bin 1;
n2 atoms are placed in bin 2, and so on up to nM atoms in bin M. The
quantities
pi = ni N
represent the probabilities of nding an atom in a particular bin. The
number of ways a specic conguration can be realized (where conguration means nding n1 atoms in bin 1, n2 atoms in bin 2, and so
on) is, from elementary probability theory, given by the multinomial
coefcient
W ({n1 , n2 , . . . , nM }) =
N!
.
n1 n2 nM
k
ln W ({ni }),
N
S = -k pi ln pi .
i =1
5.2 Frustration and rugged energy landscapes. Rugged energy landscapes are formed whenever there is a lack of good low energy
conformations. Situations of this type arise when the system of atoms
108
Figure for Problem 5.2. Nonfrustrated and frustrated squares: Atoms (vertices) are
connected by bonds (sides) with bond (coupling) strengths as indicated. (a) Nonfrustrated square in which all couplings are positive. (b) Frustrated square in which
there is one negative coupling and three positive couplings.
kT
Problems
109
6
Stress and Pheromone Responses
in Yeast
112
Figure 6.1. Signal receptors: Each receptor has an extracellular region, a transmembrane segment, and an intracellular (cytosol) portion. (a) Single-chain, singlepass receptor; (b) two-chain, single-pass receptor; (c) dimeric arrangement of
two-chain, single-pass receptors; (d) trimeric arrangement of single-chain, singlepass receptors; and (e) single-chain, seven-pass receptor. The N-terminal is usually
located on the extracellular side and the C-terminal on the cytoplasmic side in the
single-chain and seven-pass receptors. In the double-chain receptors, the uppermost
side is N-terminal and the lower side is C-terminal.
113
114
115
Figure 6.3. Enzymatic actions of protein kinases and protein phosphatases: (a) A
protein kinase catalyzes the transfer of a phosphoryl group to its substrate using
ATP as a donor. (b) A protein phosphatase catalyzes the removal of a phosphoryl
group from its substrate using ADP as an acceptor.
116
117
Figure 6.5. Signaling activities of proteolytic enzymes: (a) A protease cleaves the
tether, detaching the ligand from the outer leaet of the plasma membrane allowing it to function as a diffusible ligand. (b) A protease cleaves the polypeptide chain
just below the plasma membrane, allowing the cytoplasmic portion of the protein
to function as a downstream signaling element. (c) A protease degrades a scaffold
protein, freeing up the signaling protein to convey a signal to its downstream substrate. (d) A protease degrades a signaling protein shortening its lifetime and terminating signaling.
a message as shown in Figure 6.5(c). Proteolytic processing can do the opposite and turn off signaling. If a signaling protein rather than a scaffold is
tagged for proteolytic processing as indicated in Figure 6.5(d), the signal
proteins lifetime will be reduced and its signaling activity will be damped
down.
118
The most commonly occurring end points are those that contact and regulate the transcription machinery. Proteins that convey regulatory instructions to the transcription machinery are called transcription factors.These are
the last or furthest downstream signaling elements in the various stress and
pheromone pathways. These proteins bind to specic DNA sequences
located in noncoding regions of genes and as a consequence inuence
transcription either positively or negatively. In the former case by makig it
easier to carry out transcription and in the latter case by making it more
difcult to do so.The noncoding gene regulatory regions contain binding sites
for elements of the xed infrastructure to bind and also sites for regulatory
proteins to attach. The control regions are known as promoters and the
sequence-specic control points where transcription factors bind and come
together to regulate transcription are known as responsive elements (REs).
119
120
CaMK
CMGC
Family
Protein kinase A (PKA)/Protein
kinase G (PKG)
Protein kinase C (PKC)
Protein kinase B (PKB), Akt, RAC
G protein-coupled receptor kinase
(GRK)
Ribosomal S6 kinase (S6K)
Phosphoinositide-dependent protein
kinase-1 (PDK1)
CaM kinase (CaMK)
AMP-dependent protein kinase
(AMPK)
Cyclin-dependent kinase (CDK)
Mitogen-activated protein kinase
(MAPK)
Glycogen synthase kinase-3 (GSK-3)
Casein kinase-2 (CK2)
MEK
PIKK
Comments
Cyclic nucleotide-regulated; basic
amino acid-directed
Lipid-, calcium-regulated
Lipid-regulated
Lipid-, calcium-regulated
Lipid-regulated
Lipid-regulated
Ca2+/calmodulin-regulated; Basic
amino acid-directed
Activated when AMP:ATP is
elevated; basic amino acid-directed
Proline-directed
Substrate of MEKs; proline-directed
Principal substrate of PKB; dual
specicity; proline-directed
Uses both ATP, GTP; Acidic amino
acid-directed; dual specicity
Dual specicity; highly specic
Highly specic
Highly specic
Glutamine-directed
Proline-directed
121
kinases typically can act on more than one kind of target kinase; that is,
they have broader substrate specicity than the rst group of rather narrowly acting kinase kinases, and consequently have their own name. An
example of this kind of broader acting kinase kinase is phosphoinositide
dependent kinase (PDK). This signaling molecule acts upstream of many,
if not most members of the Protein kinase A, G, and C (AGC) group.
122
123
ing S/T residues lying within proline-rich regions. The main exceptions to
this rule are the CK2s. These kinases prefer the anking amino acids to be
acidic. Finally, phosphoinositide 3-kinase related kinases (PIKKs) are either
glutamine or proline directed, as listed in Table 6.1.
124
125
126
Figure 6.7. Pheromone response pathway: (a) Pheromones initiate signaling when
they attach to the extracellular ligand-binding portion of the G protein-coupled
receptor (GPCR). Receptor-binding triggers dissociation and activation of the Ga
and Gbg subunits of the G-protein. The Gbg subunit activates the Ste20 kinase and
interacts with the Ste5 scaffold protein resulting in activation of signaling through
a MAP kinase cascade, leading to activation of Ste12 and the subsequent transcription of genes responsive to pheromone signals, and of Far1, a mediator of cell
cycle arrest (in G1). (b) Far1 is an adapter protein and serves as an adapter between
Gbg and the Cdc24 GEF at the plasma membrane.
127
128
kinase module, along with Ste11 and Ste7, and is activated. It then
translocates back to the nucleus. There it activates the transcription factors
Ste12.
The Fus3 MAP kinase also phosphorylates and thus activates the Far1
adapter protein. This protein has several functions. It is a mediator of cell
cycle arrest (in G1) as indicated in Figure 5.7. Like Ste5, it shuttles between
different subcellular locations. It shuttles the Cdc24 GEF to the plasma
membrane and mediates formation of a complex containing several proteins at the Gbg location where the pheromone signals are strongest. This
action makes the place wherein the cytoskeleton reorients itself in preparation for bud formation.
Figure 6.8. The high osmolarity glycerol (HOG) response pathway: Two receptors,
Sln1 and Sho1, signal through a common MAP kinase cascade and downstream
Hog1 and Msn2/Msn4 transcription factors to inuence the expression of HOGresponsive genes.
129
external osmolarity, the yeast cells increase glycerol synthesis and decrease
glycerol permeability, thereby increasing their internal osmolarity and
restoring a correct osmotic gradient for water uptake.
The HOG pathway has two distinct osmosensors, Sho1 and Sln1. Sho1
and Sln1 (along with Ypd1 and Ssk1) operate in place of the GPCR and
associated G proteins that sense and transduce pheromone signals. Both
Sho and Sln1 signal through components of the HOG MAP kinase core
module resulting in the activation of the Hog1 MAP kinase. The central
element in the kinase core, Psb2, doubles both as the kinase kinase and as
the scaffold protein for the module. Signals conveyed into the cell through
the Sln1 receptor utilize Ssk2 or Ssk22 as the kinase kinase kinase, while
Sho1 signals are routed via the Ste11 kinase kinase kinase. The last element
in the MAP kinase core, Hog1, like Fus3, shuttles back and forth between
the cytoplasm and nucleus. In response to elevated osmotic stress, the
kinase is phosphorylated by Psb2 and becomes concentrated in the nucleus
along with the Msn2 and Msn4 transcription factors.
The second osmosensor, Sln1, is unusual for a eukaryotic sensor. It is a
histidine kinase, a kind of signal protein that is common in bacteria, but is
not only uncommon in eukaryotes, but also seems to be absent all together
in animals. These systems are often called histidine-aspartate (His-Asp)
phosphorelays because they involve the transfer, or relay, of phosphoryl
groups between histidine and aspartate residues. The mechanistic details of
how His-Asp systems operate in bacterial cells will be a main focus of the
next chapter.
130
Figure 6.9. General stress response pathway: Protein kinase A, the core signaling
element, is stimulated by Ras-mediated internal stress signals and by GPCR/G
protein transduced glucose signals, both acting through adenylyl cyclase/cAMP
intermediaries.
element in the internal pathway is the heat shock protein Hsp70 introduced
in the last chapter. Under low pH conditions the number of denatured proteins increases. The Hsp70 proteins must deal with this situation and are
not available to signal to the Ras GEF, Cdc25.
The G protein-coupled receptor (GPCR) functions as a sensor of glucose,
the preferred sugar for yeast cells. In response to the presence of glucose it
stimulates the production of cAMP by adenylate cyclase. These molecules
function as second messengers and have as their cellular role the activation
of protein kinase A (PKA). In the yeast this protein has a pair of regulatory subunits and a pair of catalytic subunits. It is activated when two cAMP
molecules bind to each regulatory subunit. In response, the catalytic subunits dissociate from the regulatory subunits and become activated. The
glucose-activated pathway leading from a GPCR is not the only path to
activation of PKA. Alternatively, Ras may stimulate cAMP production in
response to stress conditions, again leading to activation of PKA.
Activated PKA is a negative regulator of Msn2p and Msn4p, transcription factors (TFs) that bind to the STRE. Under good (logarithmic) growth
conditions these TFs are localized in the cytoplasm, but migrate to the
nucleus when glucose conditions are degraded. The import and export of
Msn2p and Msn4p from the nucleus is regulated by PKA activity and also
independently by signals from the TOR pathway (to be discussed next). The
131
overall mechanism is that when growth conditions are poor and the cell is
subjected to stressful conditions the Msn2p and Msn4p proteins accumulate in the nucleus, bind to the STREs, and stimulate transcription of genes
whose protein products protect the cell.
132
133
Figure 6.11. Regulation of translation of 5TOP genes through the TOR signaling
pathway: When growth conditions are favorable TOR is active and phosphorylates
the S6K kinase, which in turn phosphorylates the ribosomal factor S6 thereby
stimulating transcription of mRNAs bearing a 5TOP sequence in their upstream
untranslated (regulatory) region (UTR).
134
135
the signaling activities of many, if not most, signaling proteins in the cell.
The predominant means of movement for signaling proteins is passive
diffusion.
Particles diffuse from one location to another through random movements known as Brownian motion. Brownian motion is named after the
English botanist Robert Browning who in 1826 studied and puzzled over
the zig-zag movements of dust particles suspended in a uid that he
observed using a light microscope. Albert Einstein provided an explanation
of this phenomenon in 1905. In his analysis, Einstein showed that Brownian motion arises from random collisions of the dust particles with the molecules of the uid.
The cell interior is an aqueous medium lled with ions, biomolecules,
a cytoskeleton, and organelles. The speed at which a particular signaling
molecule can diffuse depends on three factors:
viscosity of the medium,
binding, and
crowding effects.
Diffusive motion is slower than inertial motion. In diffusion, the medium
exerts a drag or frictional force on the molecules that slow them down. The
difference between inertial and diffusive motion is reected in the mean
square displacements x2 produced by each kind of motion, namely,
x 2 =
m
kT 2
t , t <<
,
m
a
(6.1)
x 2 =
m
2kT
t , t >> .
a
a
(6.2)
and
kT
.
a
(6.3)
136
This expression formalizes the notion that as the viscosity (friction) goes
up the mobility represented by the quantity D goes down. The cellular
medium is viscous but not excessively so. Small and moderately sized biomolecules diffuse about four times slower in the cytosol as they would in a
pure water medium.
As might be expected the larger the molecule the slower it will diffuse.
This aspect of diffusive motion is encapsulated in the StokesEinstein
relationship:
a = 6 phr0 ,
(6.4)
Protein Phosphatases
Cohen PTW [2002]. Protein phosphatase 1Targeted in many directions. J. Cell Sci.,
115: 241256.
137
Janssens V, and Goris J [2001]. Protein phosphatase 2A: A highly regulated family
of serine/threonine phosphatases implicated in cell growth and signaling.
Biochem. J., 353: 417439.
Shuttling
Elion EA [2001]. The Ste5p scaffold. J. Cell Sci., 114: 39673978.
Mahanty SK, et al. [1999]. Nuclear shuttling of yeast scaffold Ste5 is required for
its recruitment to the plasma membrane and activation of the mating MAPK
cascade. Cell, 98: 501512.
Van Drogen F, et al. [2001]. MAP kinase dynamics in response to pheromones in
budding yeast. Nature Cell Biol., 3: 10511059.
138
Diffusion
Luby-Phelps K [2000]. Cytoarchitecture and physical properties of cytoplasm:
Volume, viscosity, diffusion, intracellular surface area. Int. Rev. Cytol., 192: 189221.
Verkman AS [2002]. Solute and macromolecular diffusion in cellular aqueous
compartments. Trends Biochem. Sci., 27: 2733.
FRAP
Lippincott-Schwartz J, Altan-Bonnet N, and Patterson GH [2003]. Photobleaching
and photoactivation: Following protein dynamics in living cells. Nature Cell Biol.,
5: S7S14.
Problems
6.1 Diffusion rates of various biomolecules can be measured inside the cell
using GFPs in a technique called uorescence recovery following photobleaching (FRAP). In this approach, a region inside the cell is subjected to intense laser light, which photobleaches any uorescence in
that region. GFPs located outside the exposed region then diffuse back
into the bleached region, and this movement is studied using low intensity laser light.
What is the hydration radius of a GFP molecule in water at a thermal
energy kT = 0.6 kcal/mol? Use the StokesEinstein relationship, Eq.
(6.4) together with Eq. (6.3) assuming viscosity h = 0.01 P = 0.01 g/cm
s, and a GFP diffusion coefcient D = 87 mm2/s. How long will it take
for the GFP proteins to diffuse 10 microns? Hint: Note from Eqs. (6.2)
and (6.3) that
x 2 = 2 Dt .
(This result holds in one dimension. In two dimensions, a factor of 4
appears in place of the factor of 2, and in three dimensions a factor of
6 appears.)
7
Two-Component Signaling Systems
Bacteria such as Escherichia coli are able to sense their external environments and, in response to changes in these conditions, alter their physiology. Lifetimes of bacterial proteins are generally short, and bacteria alter
their metabolism and reproductive strategies by turning on some genes and
turning off others. The corresponding signal pathways start at the plasma
membrane, where sensing takes place, and terminate at DNA control
points, where transcription is initiated. The pathways in bacteria are generally short and typically involve two core elements, a membrane-bound
sensor and signal transmitter, and a receiver that establishes contact with
the transcription apparatus.
Several representative examples of bacterial two-component signaling
pathways are listed in Table 7.1. Two kinds of signaling pathways are presented. The rst kind of pathway is represented in the table by the chemotaxis pathway. This pathway controls a piece of xed infrastructure in the
cell directly. In the case of chemotaxis, it controls the operation of the
agellar motor and terminates at a motor control point. The second kind
of pathway, represented by all other entries in the table, regulates gene
expression and by that means controls the xed infrastructure too. These
pathways terminate at DNA promoters, and the second components, the
response regulators, serve as transcription factors. Like the yeasts discussed
in the last chapter, bacteria cannot control their environments and so must
continually sense and respond to changes in their surroundings by altering
their patterns of gene expression.
Bacteria are not the only organisms that use two-component systems.
Plants use these systems to respond to hormones and to light. In the rst
part of this chapter, the bacterial chemotactic system will be examined in
detail. Binding and catalysis will be discussed and the notion of a linear
pathway, introduced in the last chapter, will be broadened to include
feedback. These explorations will be followed by a discussion of twocomponent signaling in plants.
139
140
Response regulator
Tar/CheA
EnvZ
FixL
CheY, CheB
OmpR
FixJ
KinA, KinB
NarX, NarQ
NtrB
Spo0A, Spo0F
NarL, NarP
NtrC
PhoR
PhoB
Function
Regulates chemotaxis
Osmoregulation; controls porin gene expression
Regulates nitrogen xation genes in symbiotic
bacteria
Regulates stress-induced sporulation gene expression
Regulates nitrate/nitrite metabolism gene expression
Nitrogen assimilation; controls glutamine synthetase
gene expression
Phosphate uptake gene expression
141
Figure 7.1. Phosphotransfer systems: (a) Two-component system; and (b) phosphorelay. The dark circles in the units symbolize histidine and aspartate residues
serving as binding sites for phosphoryl groups. Phosphotransfer steps are denoted
by squared-off arrows accompanied by circled Ps.
polypeptide chain. The rst is the receiver. It possesses a conserved aspartate residue that binds, or receives, the transferred phosphoryl group. The
second part of the response regulator chain is the output unit. Once fully
activated, the response regulator diffuses to, and establishes contact with,
the transcription machinery.
The second part of Figure 7.1 illustrates the operation of a typical phosphorelay. There are two additions to the system depicted in the rst part of
the gure. The sensor unit is more complex; it now becomes a hybrid sensor,
containing an extra module, an aspartate-bearing receiver functionally
identical to the receiver unit in a response regulator. Sandwiched between
the sensor unit and the response regulator is a small histidine phosphotransfer (HPt) protein. It is functionally identical to the catalytic portion of
the transmitter unit and contains the conserved histidine. As a result of
these two additions, the two-step His-Asp phosphotransfer becomes a fourstep His-Asp-His-Asp phosphorelay.
142
Figure 7.2. Domain organization of the CheA histidine kinase: A number of conserved residues characterize the histidine kinases. The conserved residues and their
surrounding 5 to 12 amino acid residues are known as homology boxes. The conserved residues are used, in one-letter amino acid code form, to name the boxes.
Four of these, the N, G1, F, and G2 homology boxes are involved in forming the
cleft that holds the ATP molecule in position. The fth, the H box, contains the conserved histidine residue involved in phosphotransfer.
Function
H domain; contains the conserved histidine (H), the site of autophosphorylation
Regulatory domain; binds CheY and CheB
Dimerization domain; binds CheA
Kinase domain; contains the asparagine (N), phenylalanine (F), and glycine-rich
(G1 and G2) boxes; binds ATP
Interaction domain; binds receptors and CheW
143
Figure 7.3. Structure of the CheA dimer: Shown are the P3 dimerization domain,
P4 kinase domain and P5 regulatory domains arranged in a dimer. The gure was
prepared using the Protein Explorer with atomic coordinates as determined by Xray crystallography and deposited in the PDB le under accession code 1b3q.
As noted above, the CheA domain organization is not the only one found
in histidine kinases. A second, rather common organization is for the H box
to be located in the C-terminal region rather than the N terminal one. In
this mode of organization, the H box containing the conserved histidine
residue is found in close proximity to the dimerization domain and the N,
G1, F, and G2 homology boxes to form a catalytic core.
144
145
Domain structures
CheY
OmpR
FixJ
NtrC
Receiver domain
Receiver domain, winged helix-turn-helix output domain
Receiver domain, four-helix bundle output domain
Receiver domain, ATPase domain, DNA-binding domain
146
147
Figure 7.5. Tar receptor: (a) Tar monomer with labels denoting the various
regionsThe presence of several regulatory (methylation) sites in the cytoplasmic
domain is denoted. (b) Tar dimerThe basic structure is that of a homodimer that
appears as a 35-nm long helical bundle oriented perpendicular to, and passing
through, the plasma membrane. The outer, ligand-binding, portion has the form of
two four-helix bundles, one per dimer subunit. Two helices from each bundle span
the plasma membrane. The cytoplasmic portion is arranged into a four-helix bundle,
formed by two helical hairpins, one from each subunit.
148
149
150
seemingly random movements of the bacterium are biased towards attractants or away from repellents through modications in the tumbling frequency arising from the sensory input signals.
Figure 7.6. Tar chemotactic signaling pathway: The receptor (Tar) and histidine
kinase (Che A) are encoded on separate chains. CheW is a scaffold that mediates
the interactions between sensor and histidine kinase. CheY is the aspartate-bearing
response regulator that mediates the cellular response (contact with agellar
motor).
Function
Receptor
Scaffold
Histidine kinase transmitter
Aspartyl response regulator
Regulatory (phosphatase)
Regulatory (methyltransferase)
Regulatory (methylesterase)
151
152
a spatial strategy, bacteria adopt a temporal one: They determine concentration gradients by comparing concentrations at slightly different times.
This is equivalent to determining a spatial concentration gradient, since
there is translational motion of the bacterium during the measurement time
interval. The key to the temporal or time derivative method is to compare
two different quantities, each measuring in some way concentration. One
quantity is the concentration at an instant in time, represented as a percent
of receptor occupancy. The second quantity is the receptor concentration a
few seconds earlier, represented by the level of receptor methylation. There
is a lag between receptor binding and the adjustment in methylation
CheA must be phosphorylated, and then CheB must be phosphorylated
before there is an adjustment in receptor methylation. The lag between
receptor binding and methylation of a few seconds serves as a memory that
enables determination of temporal gradients.
Sensing function
ethylene
ethylene
cytokinins
cytokinins
cytokinins
osmosensor
153
as several other required residues, but lack the DNA-binding domain needed
for operation as transcription factors. A-type ARRs are found in increased
numbers shortly after a cell is exposed by cytokinins. B-type ARRs contain a
DNA-binding domain in their C-terminal regions; nuclear localization
signals (NLSs) needed for entry into the nucleus; and modules associated
with Asp response regulation and signaling. Thus, the type-B ARRs contain
all the components needed for operation as transcription factors.
Cytokinin ligands are recognized by the sensor histidine kinases CRE1,
AKH2 and AKH3. Receptor-binding activates a phosphorelay from CRE1
to an HPt protein called AHP, which translocates to the nucleus and transfers its phosphoryl group to a type B response regulator. The type B RRs
activate transcription of genes encoding Type A response regulators. The
type A RRs operating in conjunction with AHPs and histidine kinases
modulate the activities in other signaling pathways (Figure 7.7).
The ethylene-signaling pathway is erected in a way that combines histidine kinase action with MAP signaling cascades. Ethylene receptors are
histidine kinases, but these proteins do not appear to transfer phosphoryl
groups to aspartyl response regulators, but rather signal through one or
more MAP-like serine/threonine kinases. The signaling mechanisms appearing in the Arabidopsis ethylene pathway bear a striking resemblance to
those of the yeast high osmolarity (HOG) pathway discussed in the last
chapter. In the ETR1 pathway, ethylene is a negative regulator of its recep-
154
Chromophore
lmax (nm)
Distribution
Rhodopsins
Phytochromes
Cryptochromes
Retinal
Tetrapyrrole
Flavins
500
665, 730
400500
Animals
Bacteria, protists, plants
Plants, insects, mammals
tor. If the ligand is not present, the receptor is in an on state and sends
signals through the MAP3 kinase CTR1. The activated CTR1 proteins negatively regulate transcription of ethylene response genes. When ethylene is
present in the environment, the receptor state is off, and it can no longer
activate CTR1. Turning off CTR1 relieves the repression by CTR1 of transcription of ethylene response genes.
155
absorbing form is designated as Pr and the far red light form as Pfr. When
the red form (Pr) absorbs light it is converted into the far-red form Pfr, and
similarly, when the far-red form (Pfr) absorbs light it is converted to the red
form Pr. The ratio of these two forms is a measure of the amount of red
light incident on the plant compared to the amount of far red light. Direct
sunlight provides more red than far red light, but the light characteristics
change when surrounding foliage shades the plant. Then the light shifts
towards the far red. Thus, during exposure to direct sunlight Pfr will predominate in the leaves and when shaded Pr will be more abundant.
The signaling pathway leading to alterations in gene expression has been
explored in Arabidopsis. The pathway is direct and short. Using green uorescent protein it has been found that the Pfr form, but not the Pr form,
translocates to the nucleus. Upon arrival, the Pfr form interacts with a transcription factor PIF3. The interaction of Pfr with PIF3 triggered by the red
light inuences a large number of cellular signaling and metabolic pathways. As observed using microarrays, some pathways are turned on while
others are turned off (Figure 7.8a).
Phytochromes are not limited to plants. They are found in photosynthetic
bacteria and in protists (algae). Cyanobacterial phytochromes are histidine
Figure 7.8. Phytochrome and cytochrome signal transduction: (a) Phytochrome signaling, in which the active form of the light receptor Pfr acts in conjunction with the
transcription factor PIF3 to regulate the expression of light-regulated genes.
(b) Cytochrome signaling, in which expression of (blue) light-sensitive genes is
turned off under dark conditions.The blue light receptor in conjunction with the regulator COP1 translocates to the nucleus where COP1 stimulates the proteolysis of
the transcription factor HY5. Under blue light conditions the Cry1/COP1 complex
remains in the cytosol and COP1 cannot inhibit HY5-mediated transcription.
156
157
Response Regulators
Baikalov I, et al. [1996]. Structure of the Escherichia coli response regulator NarL.
Biochem., 35: 1105311061.
Birch C, et al. [1999]. Conformational changes induced by phosphorylation of the
FixJ receiver domain. Structure, 7: 15051515.
Feher VA, and Cavanagh J [1999]. Millisecond-timescale motions contribute
to the function of the bacterial response regulator protein Spo0F. Nature, 400:
289293.
Kern D, et al. [1999]. Structure of a transiently phosphorylated switch in bacterial
signal transduction. Nature, 402: 894898.
Lewis RJ, et al. [1999]. Phosphorylated aspartate in the structure of a response regulator protein. J. Mol. Biol., 294: 915.
Stock J, and Da Re S [2000]. Signal transduction: Response regulators on and off.
Curr. Biol., 10: R420R424.
Volkman BF, et al. [2001]. Two-state allosteric behavior in a single-domain signaling protein. Science, 291: 24292433.
Chemotaxis Receptors
Falke JJ, and Hazelbauer GL [2001].Transmembrane signaling in bacterial chemoreceptors. Trends Biochem. Sci., 26: 257265.
Mowbray SL, and Sandgren MOJ [1998]. Chemotaxis receptors: A progress report
on structure and function. J. Struct. Biol., 124: 257275.
Ottemann KM, et al. [1999]. A piston model for transmembrane signaling of the
aspartate receptor. Science, 285: 17511754.
West AH, and Stock AM [2001]. Histidine kinases and response regulator proteins
in two-component signaling systems. Trends Biochem. Sci., 26: 369376.
Volz K [1993]. Structural conservation in the CheY superfamily. Biochem., 32:
1174111753.
158
Problems
7.1 A signicant number of the proteins discussed in the last two chapterskinases, phosphatases, GTPases, and proteasesare enzymes.
Catalysts increase the rate of the reactions by many orders of magnitude, but are not themselves altered during catalysis. In their absence
the biochemical reactions to be catalyzed are too slow because of unfavorable mixes of positive and negative charges that have to be brought
into close proximity at the transition state long enough for the reaction
to occur. As was discussed in the chapter, the histidine kinases not only
grip and position the ATP in a favorable orientation for transfer of the
gamma phosphoryl group, they also modify the electrostatic environment. Negative charges of the phosphoryl group are countered by the
positive charges of the divalent magnesium cation; a nucleophile is
present to break bonds, and the cleft at the active site in laced with
charged residues. These activities help stabilize the transition state, and
in the process lower the activation barrier for the transition to the nal
state. This lowering is depicted schematically in the gure shown below.
Problems
159
The equation shown to the right of the gure denes the three rates
involved in the catalytic process. Kinetic equations governing the catalysis
can be constructed by applying the law of mass action. The resulting expressions in terms of enzyme and substrate molar concentrations are
d[E ]
= -k1 [E ][S] + k-1 [ES] + k2 [ES],
dt
d[S]
= -k1 [E ][S] + k-1 [ES],
dt
d[ES]
= k1 [E ][S] - k-1 [ES] - k2 [ES],
dt
where the ks are the reaction rates.
The amount of enzyme is usually far less than the amount of substrate
and is completely bound up by the substrate. As a result one can make the
steady state assumption that
d[ES]
= 0.
dt
Using the above formulas, derive the MichaelisMenton equation
V0 =
Vmax [S]
,
K M + [S]
8
Organization of Signal Complexes
by Lipids, Calcium, and Cyclic AMP
161
162
Glycolipids
Cholesterol
Lipid
Phosphatidylcholine (PC)
Phosphatidylethanolamine
Phosphatidylserine
Sphingomyelin
Phosphatidylinositol
Comments
Exoplasmic leaet
Cytoplasmic leaet
Cytoplasmic leaet, negatively charged head
group
Exoplasmic leaet, sphingosine backbone
Major role in signaling
Exoplasmic leaet, cell-to-cell recognition
Inuences uidity and membrane organization
163
164
Figure 8.2. Lipids and the lipid bilayer: (a) Membrane bilayers contain a mixture
of amphipathic lipids. Each lipid molecule has a hydrophilic (polar) head region and
a two-pronged hydrophobic tail region oriented as shown. Tails are fatty acyl chains,
hydrocarbon chains with a carboxylic acid (COOH) group at one end and (usually)
a methane group at the other terminus. In cells, the polar head of each lipid molecule is surrounded by water molecules and thus is hydrated. The density of water
molecules drops off rapidly in the hydrophobic interior. (b) Small cholesterol molecules are situated in between the larger lipid molecules. Lipids differ from one
another in the number, length, and degree of saturation of the acyl chains, and in
the composition of their head groups. The overall packing density is greater in (b)
than in (a) due to the presence of cholesterol and of lipids with straighter saturated
acyl chains. Signaling proteins (not shown) carrying a GPI anchor attach to the exoplasmic leaet while signaling proteins bearing acetyl and other kinds of anchors
attach to the cytoplasmic leaet. These proteins congregate in cholesterol and
sphingolipid-enriched membrane compartments.
The plasma membranes of eukaryotes are not uniform, but rather contain
several kinds of lipid domains, each varying somewhat in its lipid composition. Compartments enriched in cholesterol and/or sphingolipids contain
high concentrations of signaling molecules: GPI-anchored proteins in their
exoplasmic leaet and a variety of anchored proteins in their cytoplasmic
leaet. Two kinds of compartmentscaveolae and lipid raftsenriched in
cholesterol and glycosphingolipids, are specialized for signaling.
Caveolae (little caves) are detergent-insoluble membrane domains
enriched in glycosphingolipids, cholesterol, and lipid-anchored proteins.
Caveolae are tiny ask-shaped invaginations in the outer leaet of the
plasma membrane. They play an important role in signaling as well as in
transport. Caveolae may be at, vesicular, or even tubular in shape, and may
be either open or closed off from the cell surface. They are detergent insoluble and are enriched in coatlike materials, caveolins, which bind to cholesterol. Cholesterol- and sphingolipid-enriched microdomains can oat
within the more diffuse lipid bilayer.
The second kind of cholesterol- and sphingolipid-enriched compartment
is a lipid raft. It does not have a cave-like shape and does not contain caveolins, but instead is rather at in shape. The uid and detergent-insoluble
properties of both the rafts and the caveolae arise from the tight packing
of the acyl chains of the sphingolipids and from the high cholesterol
content. The cholesterol molecules not only rigidify the compartment but
165
D4
D5
Members
Phosphatidylinositol-3 phosphate
Phosphatidylinositol-3,4 biphosphate
Phosphatidylinositol-3,5 biphosphate
Phosphatidylinositol-3,4,5 triphosphate
Phosphatidylinositol-4 phosphate
Phosphatidylinositol-4,5 biphosphate
Phosphatidylinositol-5 phosphate
Designation
PtdIns(3)P
PtdIns(3,4)P2
PtdIns(3,5)P2
PtdIns(3,4,5)P3 or PIP3
PtdIns(4)P
PtdIns(4,5)P2 or PIP2
PtdIns(5)P
also facilitate the formation of signaling complexes and the initiation of signaling by them.
166
167
168
Figure 8.6. PI3 kinase domain structure: (a) Classes of catalytic subunits. (b) Examples of regulatory subunits that associate with Class IA subunits of PI3 kinases.
Abbreviations: Adapter-binding (AB) domain; Ras-binding (RB) domain; Src
homology domain-3 (SH3); Src homology domain-2 (SH2); BCR-homology GTPase
activating (BH) domain.
169
Figure 8.7. Signaling through PI3K: Ligand growth factor receptor binding
stimulates the dissociation of the regulatory and catalytic subunits of PI3K from
each other. The catalytic subunit phosphorylates the PIP2 proteins at the 3 position,
thereby making PIP3, which then diffuses to and binds PDK1 and protein kinase B.
170
Domain name
C1
C2
FYVE
PH
PTB
Phosphotyrosine-binding
SH2
Src homology-2
Description
~50 amino acid residues; binds DAG
~130 amino acid residues; binds acidic
lipids
~70 amino acid residues; binds PI3P
~120 amino acid residues, binds PIP3
headgroup, PIP2 and its headgroup; binds
proteins
~100 amino acid residues; binds
phosphorylated tyrosine residues; binds
phospholipids in a weak nonspecic manner
~100 amino acid residues; binds
phosphorylated tyrosine residues; binds
phospholipids
phosphotyrosine binding domains. These domains possess phospholipidbinding properties, and for that reason have been included in Table 8.3.
171
in cells whose membranes are excitable. Whenever the membrane is depolarized the ion channels open allowing calcium ions from the extracellular
spaces to diffuse through and enter the cell. The second kind of membrane
signal molecule is the ligand-gated receptor such as the G protein-coupled
receptor. When a ligand binds the GPCR receptors, phospholipase C is activated. As discussed earlier in this chapter, PLC hydrolyzes PIP2 to IP3 and
DAG. IP3 diffuses over to, and binds to, IP3 receptors located in the ER.
This event serves as a release signal, resulting in the movement of Ca2+ out
of the stores and into the cytosol.
The duration of a calcium signal is short. Intracellular calcium levels are
restored to their base values fairly rapidly. Buffering agents bind calcium
ions before they can diffuse appreciably from their entry point. Free
calcium path lengths, the distance traveled by calcium ions before being
bound, average less than 0.5 m, which is far smaller than the linear dimensions, 10 to 30 m, of typical eukaryotic cells. In addition to being buffered,
ATP-driven calcium pumps located in the plasma membrane rapidly
remove calcium ions from the cell, and other ATP-driven pumps transport
calcium back into the intracellular stores. The take-up of calcium by buffers
along with its rapid pumping out of the cytosol and into the stores produces
a sharp localization of the signaling both in space and time.
172
Figure 8.8. Solution NMR structure of calmodulin, free and bound to calcium:
(a) Calcium-free calmodulin consisting of an (upper) N-terminal domain and a
(lower) C-terminal domain. (b) Ca2+4-bound calmodulin. The four calcium ions are
depicted as dark gray spheres. The two prominent hydrophobic patches that are
exposed in this more open conformation are bound by W7 molecules shown in a
space-lled model. The gure was prepared using Protein Explorer with atomic
coordinates deposited in the PDB under accession numbers 1dmo (a) and 1mux (b).
173
in cAMP resulting in its degradation to inert 5AMP. They also carry out
the same operation in cGMP to yield inert 5GMP. The PDEs terminate
second messenger signaling. They modulate these signals with regard to
their amplitude and duration, and through rapid degradation restrict the
spread of cAMP to other compartments in the cell.
174
Table 8.4. Members of the AGC family of serine/threonine kinases: Different geneencoded isozymes and alternatively spliced isoforms are listed in column 3. Second
messengers required for the activation of the kinases are listed in column 4.
AGC kinase family
Protein kinase A
Protein kinase B
Protein kinase C
Classical
Novel
Atypical
Structure
2 regulatory subunits;
2 catalytic subunits
Single chain; PH,
catalytic, regulatory
domains
Single chain; catalytic,
regulatory domains
Single chain; catalytic,
regulatory domains
Single chain; catalytic,
regulatory domains
Forms
Regulation
cAMP
PKC-a, PKC-b1,
PKC-b2, PKC-g
PKC-d, PKC-e,
PKC-h, PKC-q
PKC-z, PKC-i,
PKC-l
Ca2+, DAG,
phosphatidylserine
DAG,
phosphatidylserine
those belonging to the AGC family. Three subfamilies of AGC kinases are
included in Table 8.4 along with the second messengers involved in their
activation. The kinases sequester their catalytic activities within a catalytic
domain (or subunit) and similarly combine their regulatory activities into
one or more regulatory domains (or subunits). Some of the kinases possess
a separate lipid-binding PH (Pleckstrin homology) domain, while others
incorporate lipid-binding structures such as C1 and C2 domains into their
regulatory regions.
The kinases all have a common structure and similar modes of activation. Second messengers and upstream kinases activate them. Binding of
the second messengers to PH domains and regulatory motifs induces the
movements of the kinases to the plasma membrane near the sites of second
messenger release. This step is followed by phosphorylation by an upstream
kinase. In the case of protein kinase B and protein kinase C the upstream
kinase kinase has been identied. It is called phosphoinositide-dependent
kinase-1 (PDK1). This enzyme may also be the one responsible for activating protein kinase A. In all cases, the role of the upstream kinase is
to catalyze the transfer of a phosphoryl group to a crucial residue situated
in the activation loop of the AGC kinase. When this occurs the amino
acid residues involved in catalysis are unblocked and can carry out their
functions.
175
Figure 8.11. Insulin signaling through protein kinase B: (a) In the absence of PKB
activity GSK3 inhibits the stimulation of glycogen synthesis by glycogen synthase
(GS). (b) When PKB is activated it binds to and prevents GSK3 from inhibiting GS.
(c) In the absence of PKB activity GSK3 inhibits the initiation of protein synthesis
by eIF2B. (d) When PKB is activated it binds to and prevents GSK3 from inhibiting eIF2B.
176
Figure 8.12. Survival signaling through protein kinase B: (a) In the absence of PKB
signaling Bad translocates from the cytoplasm to the mitochondria where it promotes apoptotic responses. (b) When PKB is activated it phosphorylates Bad, which
then binds to a 14-3-3 protein and becomes immobilized in the cytoplasm. (c) In the
absence of PKB-signaling, the Forkhead (FH) transcription factor translocates from
the cytoplasm to the nucleus where it promotes the expression of proapoptotic
genes. (d) When PKB is activated it phosphorylates FH, which then binds to a 143-3 protein and becomes immobilized in the cytoplasm.
177
Figure 8.13. Structure of protein kinase C and protein kinase B: (a) Protein kinase
C familiesConventional (c), novel (n), and atypical (a) protein kinase C proteins.
(b) Protein kinase B and PDK1 proteins. Abreviations: pseudosubstrate (PS); Pleckstrin homology (PH); octicopeptide repeat (OPR).
178
179
ligand-gated ion channels (discussed in Chapter 18), and other membranebound signaling proteins. Other PKCs are localized to the nucleus or to the
mitochondria or to other cellular compartments where they phosphorylate
their substrates.
180
181
Figure 8.15. PKA signaling through AKAPs regulating ion channel openings:
Binding of a neuromodulator to a G protein-coupled receptor initiates signaling in
the neuron. The activated Ga subunit stimulates production of cAMP by adenylyl
cyclase. The cAMP molecules bind to the regulatory subunits of protein kinase A.
In response, the regulatory subunits dissociate from the catalytic subunits, which
then phosphorylate the ion channel, overcoming its inhibition (closed state) by the
protein phosphatases. After a time, the protein kinases are no longer able to maintain the channel in its phosphorylated state and the ion channel closes again (not
shown).
182
with high afnity. The desirable property of high afnity is combined with
another important property, substrate specicity, through use of an AKAP.
The AKAP79 situates the three signaling proteins near their targets: plasma
membrane receptors and ion channels. The enzymatic functions of these
proteins are disabled by their association with the anchoring protein.
Ca2+/CaM supplies the on signal for these enzymes. When this complex
binds protein kinase C and calcineurin, the enzymes detach, and diffuse to
and phosphorylate/dephosphorylate their nearby targets. Similarly, cAMP
binding will enable protein kinase A to detach from the AKAP79 and phosphorylate its targets.
In the brain, protein kinase A participates in the learning pathway
along with several other protein kinases. The main endpoint of the learning pathway is the nucleus where kinases functioning as transcription
factors regulate gene expression. Signaling interactions of a short term
nature involving the regulation of ion channels, and long term natureproducing changes in gene expression, are central to brain function. They
will be examined in Chapter 21.
183
Figure 8.16. Crystal structure of the conserved core of the catalytic subunit of
protein kinase A: The core unit consists of a small lobe, a linker and a large lobe.
The small lobe contains a ve-stranded antiparallel beta sheet and a conserved helix
(the C helix). The large lobe consists mainly of helices plus a grouping of four beta
strands at the bottom of the active site cleft. The phosphoryl groups covalently
attached at Thr197 and Ser338 are depicted as space-lled model. The ATP molecule is also shown with a space-lled representation. The gure was prepared using
Protein Explorer with atomic coordinates deposited in the Protein Data Bank
(PDB) under accession code 1ATP.
184
Calcium/Calmodulin
Berridge MJ, Lipp P, and Bootman MD [2000]. The versatility and universality of
calcium signaling. Nature Revs. Mol. Cell Biol., 1: 1121.
Carafoli E [2002]. Calcium signaling: A tale for all seasons. Proc. Natl. Acad. Sci.
USA, 99: 11151122.
Chin D, and Means AR [2000]. Calmodulin: A prototypic calcium sensor. Trends
Cell Biol., 10: 322328.
Adenylyl Cyclases
Cooper DMF, Mons N, and Karpen JW [1995]. Adenylyl cyclases and the interaction between calcium and cAMP signaling. Nature, 374: 421424.
Hurley JH [1999]. Structure, mechanism, and regulation of mammalian adenylyl
cyclase. J. Biol. Chem., 274: 75997602.
Taussig R, and Gilman AG [1995]. Mammalian membrane-bound adenylyl cyclases,
J. Biol. Chem., 270: 14.
Cyclic AMP
Houslay MD, and Milligan G [1997]. Tailoring cAMP-signaling responses through
isoform multiplicity. Trends Biochem. Sci., 22: 217224.
Rich TC, et al. [2001]. A uniform extracellular stimulus triggers distinct cAMP
signals in different compartments of a simple cell. Proc. Natl. Acad. Sci. USA, 98:
1304913054.
Schwartz JH [2001]. The many dimensions of cAMP signaling. Proc. Natl. Acad. Sci.
USA, 98: 1348213484.
Problems
185
PTEN
Yamada KM, and Araki M [2001]. Tumor suppressor PTEN: Modulator of cell signaling, growth, migration and apoptosis. J. Cell. Sci., 114: 23752382.
Anchoring Proteins
Colledge M, and Scott JD [1999]. AKAPs: From structure to function. Trends Cell
Biol., 9: 216221.
Feliciello A, Gottesman ME, and Avvedimento EV [2001]. The biological function
of A-kinase anchoring proteins. J. Mol. Biol., 308: 99114.
Mochly-Rosen D, and Gordon AS [1998]. Anchoring proteins for protein kinase C:
A means for isozyme selectivity. FASEB J., 12: 3542.
Protein Kinases
Huse M, and Kuriyan J [2002]. The conformational plasticity of protein kinases. Cell,
109: 275282.
Taylor SS, et al. [1999]. Catalytic subunit of cyclic AMP-dependent protein
kinase: Structure and dynamics of the active site cleft. Pharmacol. Ther., 82:
133141.
Problems
8.1 As discussed in the chapter, biological membranes can be described as
uids in which lipids and proteins freely diffuse within the plane of the
membrane. Singer and Nicholson [Singer SJ and Nicholson GL [1972].
The uid mosaic model of the structure of cell membranes. Science, 175:
720731] presented the basic features of this picture in a 1972 paper
that appeared in Science. The membranes are not homogeneous structures but rather are organized into domains, each characterized by a
somewhat different mix of lipids and proteins. A typical value for the
diffusion coefcient for lateral diffusion in the plane of the lipid bilayer
in the uid phase is D = 3 10-8 cm2/s. When the cholesterol content is
186
9
Signaling by Cells of the
Immune System
The human body has three super signaling and control systemsthe
immune system, the endocrine system, and the nervous system. Each system
has a myriad of cells extending throughout the body and specialized for
signaling. Cells of the immune system communicate using cytokines; cells
of the endocrine system send out hormones and growth factors, and the
cells of the nervous system utilize neurotransmitters and neuromodulators.
The immune system, the subject of this chapter, consists of several organs,
colonies of leukocytes, and large numbers of extracellular messengers. The
immune systems job is to identify and destroy pathogens, entities that enter
the body, establish themselves in a specic locale, or niche, multiply, cause
damage, and exit. Leukocytes, the white blood cells, are highly motile, shortlived cells that move through the cardiovascular and lymphatic systems into
damaged tissues. The leukocytes kill bacterial, protozoan, fungal, and multicellular pathogens; they destroy cells infected with viruses and bacteria, and
eliminate tumor cells.
Leukocytes are highly mobile cells that can migrate from blood into
tissues and back again into blood. They respond to infections by attacking and destroying the causative agents, or pathogens. They carry out inammatory responses, innate immune responses, and adaptive immune
responses. The inammatory response to an infection involves the triggering of physiological responses such as fever and pain, redness and swelling,
and a buildup of white blood cells, the leukocytes, at the infection site. A
local environment is formed that promotes migration of leukocytes to the
infection site, the destruction of the invasive agents, and the repair of
damaged tissues. It takes several days for the adaptive immune response to
develop, and during that the inammatory response contains the infection.
The innate immune response is a phylogenetically ancient form of
defense by multicellular organisms against pathogens. It involves recognition by host cell surface receptors of molecules situated on the outer surface
of pathogens. Such molecules are characteristic of the pathogen. Because
the receptors are encoded by the host genome and thus innate, the recognition response is termed an innate immune response. Lipopolysaccharides
187
188
189
Macrophages
Neutrophils
Function
Antigen-presenting cells (APCs); sentinels
distributed throughout the body
Produce antibodies, mediate adaptive immune
responses; antigen-presenting cells
Mediate adaptive immune responses; help B
cells respond to antigens
Kill virally infected and cancerous cells
190
191
Sending cell
Receiving cell
Instructions
IL-1
IL-2
Th cells
Activated B, T cells
IL-3
IL-4
Th cells
Th2 cells
Stem cells
Activated B, T cells
IL-5
Th2 cells
Activated B cells
IL-6
Activated B cells
Stem cells
T cells
Monocytes, Th cells
B cells
Proliferation
Growth and proliferation; Ig
production
Growth and differentiation
Proliferation and differentiation;
promotes Th2 differentiation;
Ig production
Maturation, proliferation and
differentiation; Ig production
Proliferation and differentiation;
Ig production
Growth factor for pre-T, B cells
Growth
Inhibits cytokine production
Growth and proliferation
Th1 cells
IL-7
IL-9
IL-10
IL-11
IL-12
Main role(s)
Mediates the innate immune response to
bacterial pathogens; mediates
inammatory responses and stimulates
lymphocytes
Mediates adaptive immune responses of
growth, proliferation, and death
(apoptosis)
Key mediators of the adaptive immune
response; regulates and coordinates
leukocyte activities; functions as
leukocyte-specic growth hormones,
promoting growth and differentiation
Mediates antiviral and antitumor responses,
and promotes the adaptive immune
response
Leukocyte chemoattractants
192
Figure 9.1. Helper T cell development: The rst steps in the specication involve
interactions between the immature dendritic cells and the pathogen they have
encountered. These interactions determine the precise nature of the receptor complexes expressed on the surface of the antigen-presenting dendritic cells and the
type of cytokines secreted when they interact with the nave T cells in the secondary lymphoid tissue. Differentiation is triggered by interactions between the dendritic cell (DC) and a nave T cell (Thp). The Thp expresses IL-2, which, acting in
an autocrine manner on itself, leads to its differentiation into either a Th1 cell or a
Th2 cell. IL-12 production stimulates differentiation into Th1 cells and inhibits formation of Th2 cells. IL-4 production stimulates differentiation into Th2 cells and
inhibits formation of Th1 cells.
193
194
195
Figure 9.3. The NF-kB node: Extracellular stimuli and intracellular stresses activate upstream protein kinases, which phosphorylate the IKKs. In response, the IKKs
phosphorylate the IkB proteins, triggering either ubiquitination/degradation and/or
nucleocytoplasmic shuttling, depending on the upstream signals and subunits
present. The NF-kB proteins form dimers that translocate to the nucleus where they
stimulate transcription of NF-kB responsive genes.
removed from p105 and p100 contains a series of ankyrin repeats that
are also present in the IkB proteins and mediates their sequestration.
Ankyrin repeats are 33 amino acid residues, protein-protein interaction
modules. They are found in many signaling proteins, usually as four or more
tandem-arranged repeated copies. The chain of steps leading to mobilization of the NF-kB proteins is depicted schematically in Figure 9.3.
In the absence of activating signals, the IkB proteins bind to sites in
the Rel homology domain (RHD) of the NF-kBs, thereby masking their
nuclear localization sequences (NLSs) and forcing their retention in the
cytosol. The IkBa proteins contain nuclear export sequences (NESs) and,
when they are bound to nuclear NF-kB dimers, they promote their export
to the cytosol. The inhibition on NF-kB by the IkB proteins is relived by
phosphorylation, which induces the ubiquitin-mediated degradation of the
IkBs by the 26S proteasome (Figure 9.3). IkB proteins contain N-terminal
serines that are the substrates of IkB kinases (IKKs).
The NF-kB module turns on and then turns off. One of the genes upregulated by the NF-kB proteins encodes IkBa. Signaling through the TNF
receptor leads to increased migration of NF-kB proteins to the nucleus
where they stimulate transcription of the gene for IkBa, which once synthesized binds to NF-kB, thereby shutting down its response to TNF.
Because of the time delays inherent in the signaling pathway the NF-kB
protein activity levels move up and down over time. The other IkB subunits
do not depend on the NF-kB proteins for their transcription. The subunits
activity remains fairly constant over time, enabling them to smooth out the
oscillations in NF-kB activity brought on by the negative feedback.
196
197
Figure 9.4. Mammalian MAP kinase modules:The upstream activators fall into two
groups. (a) The ERK MAP kinase module is activated by growth and mitogenic
signals relayed to it by the Ras GTPase. (b) The JNK and p38 MAP kinase modules
are activated by proinammatory cytokines and stress signals relayed to them by
the Rho family of GTPases.
198
Figure 9.5. Structure of the TRAF domain of the TRAF2 adapter determined by
X-ray crystallography: (a) Top viewShown is a trimeric arrangement of TRAF
domains that transduce signals from a 3 : 3 arrangement of TNF receptors and
ligands. (b) Side viewThe overall shape of the signaling unit resembles a mushroom with the C-TRAF domains forming the cap and the coiled-coil domains
making up the stalk. The gure was prepared using Protein Explorer with atomic
coordinates deposited in the PDB under accession number 1ca9.
199
Figure 9.6. Signaling through the IL-1/Toll pathway: Shown are the IL-1R and its
associated IL-1RAcP receptor along with several adapter proteins, namely, MyD88,
Tollip, and ECSIT, which assist in recruiting kinases and other signaling elements
to the plasma membrane. The rst step in their activation is the recruitment of the
adapter proteins to the plasma membrane. The serine/threonine kinase IRAK (IL1R-associated kinase) and the TRAF6 adapter are then assembled at the site. These
proteins activate a number of upstream kinases collectively designated in the gure
as Tak1/Tab1/NIK, which then activate the NF-kB pathway and the JNK/p38 MAP
kinase pathways.
200
Some tissues such as the eye, testis, and parts of the central nervous system
cannot tolerate inammation. These are privileged sites that insulate themselves from immune responses. If a lymphocyte comes into contact with a
cell in any of these sites it will rapidly undergo apoptosis. Tumor cells trying
to evade the immune response use this mechanism, as well.
The TNF receptors can be divided into two groups according to the kind
of adapter proteins they use. Members of one group of TNF receptors have
TRAF-binding motifs in their cytoplasmic domain, which mediate binding
to TRAFs, while members of the second group have death domain (DD)
motifs and bind to DD-bearing adapters. Receptors belonging to the second
group are called death receptors because they convey cell suicide (apoptosis) instructions to the recipient. Upstream signaling events in these two
TNF pathways are depicted in Figure 9.7. Most TNF ligands are single-pass
transmembrane proteins with their C-terminus outside the cell and their Nterminus in the cytoplasm. They contact TNF receptors expressed on the
surface of an opposing cell. The arrangement of receptors and ligands is
3 : 3. Three receptors arranged symmetrically contact three ligands. This
signaling arrangement is facilitated by a trimeric arrangement of adapters
of the form illustrated in Figure 9.5.
201
Jak
STAT
Jak2
Jak2
STAT5a, STAT5b
STAT5a, STAT5b, STAT3
Jak1, Jak3
Jak1, Jak3
Jak2
STAT5a, STAT5b
STAT3
IL-12:
IL-12Rb1, IL-12Rb2
Jak2, Tyk2
STAT4
Class II-Interferon/IL-10:
IFNa, IFNb
IFNg
IL-10
Jak1, Tyk2
Jak1, Jak2
Jak1, Tyk2
STAT1, STAT2
STAT1
STAT1, STAT3, STAT5
202
Figure 9.8. Structure of the human growth hormone (hGH) ligand bound to two
hGH receptors: Shown are two extracellular domains of the hGH receptors bound
to a single hGH ligand. The hGH ligand binds in the cleft between the two extracellular domains of the receptors. The gure was prepared using Protein Explorer
with atomic coordinates deposited in the PDB under accession number 1hwg.
203
204
Figure 9.9. Domain organization of Jaks and STATs: (a) Janus tyrosine kinases, or
Jak proteins. (b) Signal transducer and activator of transcription (STAT) proteins.
signal. The solution to this problem of how to transduce a signal into the
cell is solved by dimerization. The combined effect of ligand binding and
dimerization alters the intracellular electrostatic environment sufciently
to stabilize a different mix of conformational states that will activate the
Jaks, triggering phosphorylation and the subsequent recruitment of the
STATs and other signaling elements to the receptors. The Jaks (tyrosine
kinases) and STATs (transcription factors) are then able to transduce the
hematopoietin signals into the cell responses.
The domain organization of the Jak and STAT proteins is presented in
Figure 9.9. As shown in part (a) of the gure, the Janus kinases possess
kinase and pseudokinase domains that face one another, hence the name
Janus kinase (Jak), named after the Roman god of gates and doorways.They
also possess a receptor-binding region that mediates their binding to recognition motifs in the cytoplasmic terminal of the cytokine receptors. Once
recruited to the receptors, autophosphorylation occurs, resulting in their
activation. The activated kinases then phosphorylate the receptors, thereby
exposing docking sites for the STATs. As depicted in part (b) of the gure,
the STATs possess a dimerization domain, an SH2 domain, a tyrosine phosphorylation site, and a DNA-binding domain. The signal transducer and
activator of transcription proteins are transcription factors, and they are
recruited to the activated signaling complex formed by the phosphorylated
cytoplasmic receptor terminals and Jaks. They bind the phosphorylated
tyrosine residues in the receptors through their SH2 domains, and they are
phosphorylated on their tyrosine residues by the Jaks. Once these recruitment and activation steps take place, the STATs are able to form heterodimer and homodimers, and translocate to the nucleus where they
stimulate transcription of their target genes.
205
206
Figure 9.11. Interferon signaling: Displayed in the gure are the main components
of the IFNa/b and IFNg pathways from the plasma membrane to the nucleus. Distinct a and b chains of each receptor jointly bind a ligand. Janus family kinases are
recruited and are activated. They create docking sites for the STATs by phosphorylating the receptors. The STATs are recruited and are phosphorylated by the Jaks.
STATs form dimers (trimers) and translocate to the nucleus where they bind to
interferon-responsive DNA transcriptional sites, the ISREs and interferm-gamma
activated sites (GASs).
The trimers translocate to the nucleus where they stimulate transcription of IFNa/b-responsive genes IFNg utilizes a different set of signal
transducers. Jak1 and Jak2 are recruited to the plasma membrane and
phosphorylate STAT1s, which form homodimers that translocate to the
nucleus where they stimulate transcription of IFNg-responsive genes.
Cytokines such as the interferons mount a multilayered response. Besides
arrest of protein synthesis and cell multiplication, they stimulate the
production of enzymes that kill (degrade) the viruses, recruit cells that
attack virus-containing cells, and organize other elements of the adaptive
immune response.
207
phoid tissues. Thus, they act to maintain homeostasis among the various
populations of lymphocytes. Stromal cells, endothelial cells, and other cells
residing in the lymphoid organs produce these signaling molecules.
Chemokines have a positive charge and bind to heparin and heparin
sulfate, negatively charged polysaccharides found within the extracellular
matrix and on the surface of endothelial cells. The heparin and heparin
sulfate molecule act as receptors for the chemokines. The chemokines
become immobilized when bound to the polysaccharide-covered substrates
and form stable gradients of chemokine concentration. The migratory
leukocytes navigate up the gradients to the target sites.
Chemokines are bound at the cell surface by G protein-coupled receptors (GPCRs), seven-pass transmembrane receptors coupled to G proteins.
Their method of transducing signals into the cell was examined briey in
the last chapter and will be discussed in detail in Chapter 12. There are four
families of chemokine receptors. These families are distinguished from one
another by their structure and by their chromosomal locations. The distinguishing structural feature is the presence of four conserved cysteine (C)
residues in specic positions. The CXC and CC families are fairly large with
several members each, while the two small families, designated as CX3C
and C, have a single known member each. The CXC family has an amino
acid located between the rst and second cysteines; in the CC family the
two cysteines are directly adjacent to one another. There is considerable
redundancy; within each ligand-receptor family, the chemokines can bind
to more than one type of receptor, and the receptors bind to more than one
type of chemokine.
Lymphoid chemokines form chemical highways within lymphoid organs
that guide the migration of lymphocytes from site to site bringing together
lymphocytes that must interact with one another in the adaptive immune
response. The B-cell attracting chemokine (BCA-1), and secondary lymphoid chemokine (SLC) are representative lymphoid chemokines. They
bind the CXCR5 and CCR7 receptors, respectively. Gradients of BCA-1
chemokines guide the entry of T cells and dendritic cells into lymphoid
organs. Similarly, gradients of SLC are crucial for compartmental homing
of B cells and T cells, helping to establish distinct territories within the secondary lymphoid organs.
208
209
the cell surface. Pathogen peptides trigger responses even though they are
only a small fraction of the total peptide fragment populationfrom 0.01
to 0.1%. Thus, the MHC must exhibit high sensitivity and selectivity of
foreign peptides while maintaining low responsiveness to self-peptides.
Different individuals express different mixes of these molecules. These
proteins lie at the core of self versus not-self recognition. The job of an
MHC is to present antigens to T cells and to NK cells. Class I MHCs interact with CD8 receptors on cytotoxic T cells and with KIR receptors on NK
cells. Class II MHCs interact with CD4 receptors on helper T cells. Class I
members bind peptides derived from molecules encountered in the cytosol
while Class II members bind peptides from molecules broken down in the
endosomes.
What the MHCs of both classes have to do is present the broadest range
of peptides possible while simultaneously satisfying the requirement that
these peptides be presented in a way that maximally stimulates TCR
responses. The minimal size of a peptide fragment needed to discriminate
between self and foreign is about 8 or 9 amino acid residues. One way of
satisfying the broadness requirement is to have several binding sites along
the MCH. However, this would violate the TCR stimulation requirement.
To maximally induce a TCR response, there should be a single peptidebinding site with each peptide of a given sequence binding to a given MHC
allele in exactly the same way. The solution is to have a single binding
pocket and a way to generate a spectrum of different MHCs utilizing a combination of conserved and nonconserved (variable region) residues in the
binding pocket.
210
Figure 9.12. T cell receptor (TCR) signal transduction pathways leading to expression of genes activated by AP-1, NF-kB, and NFAT transcription factors working in
concert: In response to ligand binding at the TCR, CD45 (a receptor tyrosine phosphatase) and CD4 activate Fyn and Lck. Fyn and Lck phosphorylate the g, d, and e
chains of the CD3 complex and the z dimer. After phosphorylation, ZAP-70 containing two SH2 domains is recruited and binds to the phosphatyrosines and is then
activated through phosphorylation by Lck. ZAP-70 then interacts with PLC-g, which
subsequently cleaves PIP2 to produce IP3 leading to the aforementioned increased
concentration of intracellular calcium. DAG activates PKC, which serves as the
upstream kinase for activation of NF-kB, while ZAP-70 acts through a RasGEF,
Ras, and a MAP kinase cascade leading to activation of AP-1. Intermediate steps
involved in activation of NF-kB by PKC have been omitted for simplicity.
cytotoxic T cells, express CD8 proteins on their surfaces. The CD8 proteins
act in concert with the T cell receptors to bind peptides derived from antigens bound to MHC-I molecules.
Several transmembrane proteins must associate with one another for efcient signaling into the cell. The upstream signaling starts with ligand
binding and association of the TCRs with their CD4 or CD8 coreceptors
and with CD3 chains (Figure 9.12). CD3 consists of several separate chains,
denoted as d, g, and e. In addition, there is a fourth kind of chain, called z.
These chains form homo- and heterodimers that associate with the a/b
chains of the TCR. The CD3 and z chains possess immunoreceptor tyrosine-based activation motifs, or ITAMs, in their cytoplasmic regions. These
enable the CD3 chains to recruit and bind Fyn and Lck protein tyrosine
kinases (protein tyrosine kinases will be discussed in detail in Chapter 11),
triggering a series of steps leading to activation of AP-1, NF-kB, and NFATresponsive genes.
Signaling through the TCR and its associated proteins culminates in the
transcription of genes-encoding receptors, signal transducers, cell adhesion
211
Ligand
Comments
CD40
CD40L
TNF superfamily
CD28/B7
CD28
CTLA-4
ICOS
PD-1
B7-1, B7-2
B7-1, B7-2
ICOSL
B7-L1, B7-L2
Ig superfamily
Positive regulator
Negative regulator
Positive regulator
Negative regulator
212
CD40/CD40L activates resting B cells and primes the dendritic cells to adequately stimulate the T cells.
The remaining receptors and ligands all belong to the CD28/B7 branch
of the immunoglobulin (Ig) superfamily. The rst of these, CD28, is constitutively expressed on the surface of most T cells. Signaling through CD28
helps determine how the T cells will differentiate. In the absence of the costimulatory signals the T cells undergo anergy, becoming nonresponsive, or
tolerant, of antigens presented on the surface of the antigen-presenting cells
(APCs). In this state the T cells are inactive. They do not proliferate nor do
they send out cytokines signals. The co-stimulator signals not only prevent
anergy but also help determine whether the activated T cell will become a
helper T cell or an effector T cell.
CD28 and CTLA-4 both bind to a pair of ligands, B7-1 and B7-2. The
CTLA-4 receptor is transiently expressed. It has a higher afnity for the B7
ligands than does CD28, and when it is expressed it shuts down the signaling.
A second pair of positive and negative regulators, ICOS and PD-1, also
appears in the table.These glycoproteins are not constitutively expressed but
are inducible through CD28 signaling and act later during immune responses.
The positive regulator, ICOS, helps promote B-T cell interactions while
PD-1, like CTLA-4, halts cytokine production and the cell cycle progression.
213
with transducing a signal from the extracellular face to the cytoplasmic face.
The receptors work together with coreceptors and with cell adhesion molecules to relay information into the cell.
In the immune systems receptors cluster and rearrange themselves.
Signals received at the cell surface help shape the clusters; the character of
the cluster in turn shapes the signal transduced into the cell interior. The
lipid bilayer is not passive but instead plays an active role in these adaptive
rearrangements. These procedures are utilized by leukocytes, by nerve cells,
and, as was discussed in Chapter 7, in the bacterial chemotactic system.
The proteins required for signaling between T cells and APCs are organized into signaling structures called immunological synapses (ISs). The term
synapse denotes a junction between cells where information is transmitted. Synapses are highly enriched in signaling molecules that not only transmit signals but also adaptively control the properties of the relayed
messages such as their strength. The term synapse originates in studies of
the transmission of signals between nerve cells. The literal meaning of the
term, coined by Sir Charles S. Sherrington in 1897, is to clasp together.
Synapses between nerve cells will be explored in Chapter 21.
Immunological synapses bring together a host of proteins required to
support sustained signaling. Among the components of the synapse are
receptors, cytoplasmic kinases, and adapters and anchors that link events at
the cell surface to the cytoskeleton. The TCRs and MHCs lie in the middle
of immunological synapse. As discussed earlier, the TCRs associate with
cytoplasmic CD3 complexes, protein tyrosine kinases such as Lck and Fyn,
and costimulatory molecules such as CD28 and CD40. Other proteins
belonging to the immunological synapse include ICAM-1 immunoglobulins
and LFA-1 intergrins, which bind one another, and members of the CD2
grouping of Ig adhesive molecules, which include CD48 (rat) and CD58
(human). This second group of mainly adhesive proteins surrounds the
central portion of the immunological synapse.
214
General References
Benjamini E, Coico R, and Sunshine G [2000]. Immunology (4th edition) New York:
John Wiley and Sons.
Janeway CA, Jr., Travers P, Walport M, and Capra JD [1999]. Immuno Biology (4th
edition) London: Current Biology Publications.
215
TRAFs
Arch RH, Gedrich RW, and Thompson CB [1998]. Tumor necrosis factor receptorassociated factors (TRAFs)A family of adapter proteins that regulate life and
death. Genes Dev., 12: 28212830.
Chung JY, et al. [2002]. All TRAFs are not created equal: Common and distinct
molecular mechanisms of TRAF-mediated signal transduction. J. Cell Sci., 115:
679688.
216
Medzhitov R, and Janeway C, Jr. [1997]. Innate immunity: The virtues of a nonclonal
system of recognition Cell, 91: 295298.
Medzhitov R, Preston-Hulburt P, and Janeway CA, Jr. [1998]. A human homologue
of the Drosophila Toll protein signals activation of adaptive immunity. Nature,
388: 394397.
TNFs
Bodmer JL, Schneider P, and Tschopp J [2002]. The molecular architecture of the
TNF superfamily. Trends Biochem. Sci., 27: 1926.
Locksley RM, Killeen N, and Lenardo MJ [2001]. The TNF and TNF receptor superfamilies: Integrating mammalian biology. Cell, 104: 487501.
Chemokines
Baggiolini M [1998]. Chemokines and leukocyte trafc. Nature, 392: 565568.
Cyster JG [1999]. Chemokines and cell migration in secondary lymphoid organs.
Science, 286: 20982102.
Rollins BJ [1997]. Chemokines. Blood, 90: 909928.
T Cell Costimulation
Lanzavecchia A [1998]. License to kill. Nature, 393: 413414.
Schwartz RH [2001]. It takes more than two to tango. Nature, 409: 3132.
Sharpe AH, and Freeman GJ [2002]. The B7-CD28 superfamily. Nature Rev.
Immunol., 2: 116126.
Viola A, et al. [1999]. T lymphocyte costimulation mediated by reorganization of
membrane microdomains. Science, 283: 680682.
Immunological Synapses
Coombs D, et al. [2002]. Activated TCRs remain marked for internalization after
dissociation from pMHC. Nature Immunol., 3: 926931.
Problems
217
Dustin ML, and Cooper JA [2000]. The immunological synapse and the actin
cytoskeleton: Molecular hardware for T cell signaling. Nature Immunol., 1: 2329.
Grakoui A, et al. [1999]. The immunological synapse: A molecular machine controlling T cell activation. Science, 285: 221227.
Krummel MF, et al. [2000]. Differential clustering of CD4 and CD3z during T cell
recognition. Science, 289: 13491352.
Lee KH, et al. [2003]. The immunological synapse balances T cell receptor signaling and degradation. Science, 302: 12181222.
Monks CRF, et al. [1998]. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature, 395: 8286.
Problems
9.1 There are two basic approaches used in theoretical studies of how signaling pathways operate: macroscopic and microscopic. The starting
point for the macroscopic approaches is the law of mass action, and for
the microscopic approach it is the principle of detailed balance. In both
classes of approaches one constructs a set of equations that captures
the salient features of the signaling process in terms of rates and other
quantities that can be related to experiment. One then follows the evolution of the system on a computer to answer questions posed by the
experimental data. In many situations, modeling a system and then simulating its behavior on a computer is the only way to see just what the
biomolecules are doing over time.
The law of mass action is a statement that the rate of a chemical reaction is proportional to the concentrations of the reactants. To illustrate
how this rule works, consider a reaction of the form
(9.1)
In the above, the rate of change of a reactant is expressed as the difference between its rate accumulation and its rate of loss through
degradation and additional/reverse reactions. Expressions of this form
can be simplied if some of the concentrations do not change appreciably over time, if some intermediates are formed rather transiently
compared to the other steps, and if the reaction only proceeds in one
direction or the other. For example, if the concentration of D does not
change over time the second term in Eq. (9.1) becomes a linear one in
which [D] is absorbed into the constant. An intermediate step, where a
218
d X i +1
dt
] = k [X ][X
p
i +1
] - ji+1 [X i+1 ],
p
i +1
(9.2)
where [Y] has been assumed to be constant and was absorbed into the
phosphatase rate constant j. Other equations can be constructed that
describe interactions between ligands and receptors, between receptors
and kinases/adapters, and between adapters and kinases. The result is
a model of each of the steps in the signaling pathway.
(a) Derive the above expression, Eq. (9.2), for the rate of change in
[Xi+1P]. This equation can be solved to determine the steady-state (ss)
concentration of Xi+1P.
(b) Show that the result, taking the concentration of the unphosporylated plus phosphorylated forms of Xi+1 to be a constant T, is
X T
.
] = [ ]
( j k ) + [X ]
p
[X
p
i +1
ss
i +1
i +1
(9.3)
This steady-state formula, Eq. (9.3), relates the stimulus [XiP] to the
response [Xi+1P]. What is the behavior of the resulting stimulusresponse curve at low [XiP], and at high [XiP]? How does this compare
to MichaelisMenten kinetics discussed in the problems for Chapter 7?
9.2 The reaction kinetics models based on the law of mass action make up
a macroscopic description of the biochemical processes taking place.
Alternatively one could employ a microscopic description that deals
with the movement and interactions at the molecular and atomic levels
rather than dealing with macroscopic quantities such as the concentra-
Problems
219
tions. The starting place for the microscopic approaches is the principle
of detailed balance, which states that the rate at which a system makes
a transition into a particular microstate from a preceding state is equal
to the rate that the system will transition back out of that particular
microstate to its predecessor. In mathematical terms the principle is
pi Pij = p j Pji ,
(9.4)
where pi is the probability that the ith microstate is occupied (i.e., the
occupation probability), and Pij is the transition probability from
microstate i to microstate j. The above expression is a statement that
the laws of physics are the same going forward or backward. In particular, the expression states that the probability that a state is occupied
times the rate at which there is a transition out of that state to another
is equal to the probability that the entered state is occupied times the
rate at which that state is exited back to its predecessor. The occupation probabilities can be dened in terms of the Boltzmann factor b:
pi =
1 -bEi
e ,
Z
(9.5)
(9.6)
In the simulations, one starts out with the system in some initial state,
then randomly picks a new microstate and calculates the energy difference DEij = Ej - Ei. If the energy difference is negative then the transition leads to a state of lower energy and the move is allowed. If the
energy difference is positive, then the transition increases the energy of
the system. One then selects a random number between 0 and 1. If that
random number is less than the calculated Pij, the transition is allowed.
Otherwise the move is rejected and another proposed transition is
selected. Note that in this approach moves that increase the energy
become possible allowing the system to transition over barriers and out
of pockets in the energy landscape. This would not be possible in situations where only energy decreasing steps were allowed.
This algorithm, known as the Metropolis algorithm, was introduced
in a landmark paper in 1953. The general method of randomly sampling
a set of moves of a microsystem according to a probability distribution
is known as the Monte Carlo method and is extensively used in several
220
10
Cell Adhesion and Motility
Movement and adhesive contacts between cells and opposing surfaces are
necessary for embryonic development and normal adult functioning of cells
in the body. During development, cells do more than grow, multiply, and
differentiate. They also segregate, migrate, and aggregate in forming tissues
and organs. In adult life, vascular endothelial cells and broblasts migrate
during wound healing, leukocytes migrate in response to infections, and
cancerous cells migrate during metastasis.
Cells establish and maintain contacts with other cells and with the extracellular matrix. These contacts stabilize the aggregates of cells and enable
them to work together to carry out their functions. Adhesive contacts
underlie axon outgrowth during development of the nervous system. Paralleling the steps taken by migrating cells, growth cones, motile sensory
structures enriched in adhesion molecules, guide the formation of connections in the nervous system.
Several different families of cell adhesion moleculesintegrins, cadherins, immunoglobulin superfamily cell adhesion molecules (IgCAMs),
and selectinsmediate adhesive contacts between surfaces in the body.
These molecules, acting as receptors and counterreceptors/ligands on
opposing surfaces, are responsible for establishing and maintaining physical contact and communication between the extracellular matrix, the cell
surface, and the cytoskeleton. These families of adhesion molecules, along
with several different kinds of diffusible molecules and their receptors, also
mediate axon outgrowth in the nervous system. Leukocyte adhesion and
mobility will be examined in the rst part of this chapter and axon outgrowth in the second.
222
Figure 10.1. Representative extracellular regions of proteins involved in cell adhesion: (a) Classical cadherins containing 5 cadherin domains N-terminal to the transmembrane segment shown in black. (b) Neural cell adhesion molecules (NCAMs)
containing 5 Ig-like domains and 2 bronectin type III (FNIII) repeats. (c) Eselectins containing an amino terminal lectin domain, an EGF domain, 6 short consensus repeats (SCRs), and a membrane proximal cleavage region.
223
work synergistically with receptors for cytokines and soluble growth factor.
They signal through many of the same pathways as growth factors and
together promote cell survival, proliferation, and differentiation. The
cytoplasmic domains of cell adhesion receptors make contact with the
cytoskeleton, and the receptors help regulate cell shape and polarization,
and cytoskeleton organization and motility. During metastasis cancer cells
utilize a similar ensemble of mechanical and adhesion receptor signaling
processes. In metastasis, cells detach from the surrounding tissue, migrate
to remote sites elsewhere in the body using the lymphatic and circulatory
systems, reattach, and then establish a colony. Signaling between the cell
surface and the nucleus coordinates the expression of specic cell adhesion
molecules at different stages in metastasis.
Cell adhesion receptors are both large and exible and because of these
properties present challenges in their study using high resolution NMR and
X-ray crystallography methods. The solution to this technical challenge is
to take advantage of their inherent modularity and characterize portions
fragments and domainsof these molecules. The resulting NMR and X-ray
crystallography data together with electron microscopy results provide a
core body of structural information on these essential proteins.
224
(b)
(c)
ligand binding. X-ray crystallography data revealing this bent shape are
presented in Figure 10.2.
Integrins and cadherins differ from most transmembrane signal transducers that transmit signals in one direction, from outside the cell inward.
Integrin and cadherin receptors transmit signals in both directionsfrom
outside the cell inward (outside-in) and from inside the cell outward
(inside-out). In outside-in signaling, binding of the integrins to the ECM
triggers changes in the pattern of gene expression. Inside-outside signals
produce changes in the integrin conformation resulting in changes in adhesiveness. Integrins tie the ECM to the cellular cytoskeleton and anchor cells
in a xed position, while cadherins are a primary element of cell-to-cell
junctions.
225
Figure 10.3. Integrin crystal structure: (a) V-shaped, or bent, closed conformation
of the aVb3 integrin. The alpha chain is shown in light gray and the beta chain in
black. (b) Complex formed between two aL inserted (I) domains (black) and a pair
of ICAM-1 ligands (light gray). The gure was prepared using Protein Explorer with
atomic coordinates deposited in the PDB under accession numbers 1jv2 (a) and
1mq8 (b).
226
proteins, are the primary ECM elements. Collagens are long ropelike linear
molecules composed of three chains arranged in a triple helix. Typical
lengths of collagens are 300 to 400 nm. Two families of adapter molecules
are present in the basal laminalaminins and bronectins. These molecules
connect ECM collagens to adhesive cellular proteins.
The extracellular matrix has considerable inuence over the metazoan
cell. Both mechanical and chemical signals are sent from the ECM to the cell
surface, and from there they are transduced into the cell interior. Integrins
have as their ligands the aforementioned ECM adapter molecules laminin
and bronectin. Binding to these proteins triggers the formation of adhesive
contacts at specic locations along the plasma membrane. Binding to
the matrix proteins stimulates integrin clustering, signaling to proteins
embedded in the plasma membrane,and to proteins located in the cytoplasm.
The cytoplasmic tails of integrin beta chains contain motifs of the form
NPxY (arginine-proline-X-tyrosine) that are recognized by phosphotyrosine-binding (PTB) domains of cytoplasmic adapter molecules, nonreceptor tyrosine kinases such as Src, focal adhesion kinase (FAK), and the actin
cytoskeleton-binding protein talin. The binding of the beta chains to talin
and other proteins establishes mechanical linkages called focal adhesions
between the ECM, the cell surface, and the cytoskeleton that facilitates clustering of integrins and the onset of signaling. Once the focal adhesions are
formed positive feedback signals to the integrin receptors stimulate their
further clustering and additional signaling from them to the focal adhesions.
The result of this two-way signaling is the tying together, or integration, of
the ECM with the cytoskeleton of the cell. The integrative properties of
these receptors led to their being given the name integrins.
227
Classical cadherins such as E-cadherin (epithelial cadherin) and Ncadherin (neural cadherin), contain about 750 amino acid residues. Their
extracellular domain consists of ve repeats of a 100-amino acid EC
domain, which extends out 250
from the cell surface (Figure 10.4). These
cadherins attach to the actin cytoskeleton by means of a linker protein
called catenin. The cytoplasmic domain of these cadherins is approximately
150 amino acid residues in size, and contains a catenin-binding site. There
are two catenin subunits. The b subunit binds to the catenin domain of the
cadherin and to the a subunit of the catenin. The a subunit, in turn, connects to the actin cytoskeleton. This cadherin-catenin complex is referred
to as a zonula adherens junction in epithelial cells and as an adherens junction in neurons. In epithelia, these junctions link cells together laterally to
permit them to form stable sheets, and they separate the apical (top) region
from the basolateral (bottom) region of each of the cells. In neurons, the
adherens junctions connect the pre- and postsynaptic cells, thereby providing mechanical stability and a means of conveying signals in both directs
across the synapse.
Cadherins operate in a homophilic manner, and have a role in maintaining tissue boundaries and stabilizing synapses. Cells that segregate into
distinct tissues express different cadherins, so that a cadherin receptor on
the surface of one cell binds to a cadherin receptor of the same type on the
surface of an opposing cell. Calcium binding is necessary for clustering, or
oligomerization, of cadherin receptors as well as for ligand binding. An
example of how this requirement might serve a useful function is provided
by N-cadherin binding in synaptic junctions. Intense synaptic activity generates long-lasting changes in synaptic transmission. Remodeling of the
synapse underlies these changes. These changes might occur through the
following sequence of steps: Intense activity depletes the supply of calcium
in the vicinity of the synapse. This lowering in the local calcium concentration weakens the links between cadherin molecules allowing remodeling
228
CD designation
Ligand
Distribution/role
Lymphocytes, endothelial
cells when inammation is
present
Recirculating leukocytes
Leukocytes
Lymphocytes
Broad distribution
Neural tissue
Platelets, leukocytes
Lymphocytes, endothelial
cells when inammation is
present
ICAM-1
CD54
ICAM-2
ICAM-3
LFA-2
LFA-3
NCAM
PECAM-1
VCAM-1
CD102
CD50
CD2
CD58
CD56
CD31
CD106
LFA-1, Mac-1
LFA-1
LFA-3
LFA-2
NCAM, collagen, heparin
PECAM-1
a4b1 and a4b7 integrins
229
group of IgCAMs are the DCC family members. Named for the protein
DCC (deleted in colorectal cancer) these are 4/6 IgCAMs. Molecules closely
related to DCC have a prominent role in the development of the nervous
system. They are found on growth cones where they bind a family of diffusible chemoattractants called netrins that guide growing axons during
embryonic development, as will be discussed later in this chapter.
The two most prominent families of IgCAMs are the NCAMs and ICAMs
(intercellular cell adhesion molecules). The ICAMs bind to integrins and
thus mediate heterophilic binding. NCAMs bind to identical receptors on
opposing cells and thus mediate homophilic binding. Besides their role in the
immune/inammatory system, these proteins play an important role during
embryonic development and in the nervous system both during development and in adult life. NCAMs are also expressed in the heart, gonads, and
skeletal muscle. Because of their role in signaling and maintaining tissue
integrity they have a prominent role in several forms of cancer.
230
Table 10.2. Selectins, cells, and structures that express them, their functions, and
ligands: Ligand abbreviations are as follows: PSGL-1 (P-selectin glycoprotein
ligand-1); MAdCAM-1 (mucosal addressin cellular adhesion molecule-1);
GlyCAM-1 (glycosylation-dependent cell adhesion molecule-1).
Selectin
Expression
L-selectin
Leukocytes
P-selectin
E-selectin
Platelets, endothelium
Endothelium
Functions
Leukocyte trafcking,
rolling adhesion
Rolling adhesion
Rolling adhesion induced
by inammation
Ligands
PSGL-1, GlyCAM-1,
MAdCAM-1, CD34
PSGL-1
PSGL-1
231
The leukocytes do not passively oat along the blood vessels, but instead
remain in contact with the cells lining the walls of the blood vessels. They
move along the walls by rolling in a controlled way. This mode of locomotion enables them to stop at the right location and then exit by crawling out
between the cells. Adhesion is critically important, enabling the leukocytes
to rst roll, and then crawl, and nally to kill the pathogens.
In an inammatory response, the physiology of the blood vessels is
altered to better enable the leukocytes to reach the site of infection. Postcapillary venules are the main locus of vascular inammatory activity during
an inammatory response. These structures, some 30 to 40 microns in diameter, consist of a layer of endothelial cells on the inside and a supporting
basement membrane on the outside. Blood ow in small vessels such as the
postcapillary venules takes place under low ow rates, reasonably high
viscosities, and small vessel diameter. Under these conditions the velocity
of the blood ow is greatest in the center of the vessel and decreases to
zero as the vessel walls are approached. During inammation the blood
vessels dilate and the overall ow rate is slowed. Red blood cells aggregate
into large assemblies called rouleaux that collect into the center of the
vessels displacing white blood cells. In response the leukocytes move from
the center of the blood vessel to the vicinity of the vessel walls. When this
happens cell surface receptors expressed on the plasma membranes of the
endothelial cells and leukocytes can engage one another to promote rolling
and then hard adhesion.
232
10.11 Slip & Catch Bonds Between Selectins & Carbohydrate Ligands
233
Figure 10.6. Energy landscape for dissociation of bonds between adhesive molecules and their ligands: Abbreviationstransition state 1 (TS1); transition state 2
(TS2); intermediate state (IS).
across the cantilever and measuring the angle and orientation of the
reected light. In an optical tweezers experiment, one uses a laser beam to
trap dielectric particles. External forces applied to the trapped particles can
be determined by monitoring the angular deection of the laser light. The
adhesive molecules are mounted on a pedestal attached to a surface and
the ligands are attached to optically trapped beads.
Experiments of several different adhesion molecule/ligand combinations
have been studied, and the results placed within an energy-landscape perspective. A representative landscape deduced from these experiments is
presented in Figure 10.6. This landscape has two prominent features. First,
it is a rugged landscape. The pockets are deep, several times the thermal
energy, resulting in the presence of at least one metastable intermediate
state. The situation shown in the gure is that of two transition states.
Second, two different outer barriers heights are presentone of these corresponds to low afnity binding and the other to high afnity binding. The
high inner barrier is responsible for high strengths seen at short time scales
and the major portion of the activation energy. The lower barriers located
further out extend bond lifetimes when forces are absent.
234
leukocytes to detach from surface tethers at the right time while maintaining good adhesion to that place on the surface at earlier times. The corresponding bonds are called slip bonds because they allow the rolling cells to
slip away. These are not the only kinds of bonds that form. An observation
often made not only of rolling leukocytes, but also migrating bacteria, is
that shear stresses promote good surface contact. The physical mechanism
underlying this phenomenon is captured by the notion of a catch bonda
bond that is strengthened by the external forces rather than weakened. The
selectins that mediate leukocyte rolling exhibit both kinds of bonds. The
catch bonds are formed rst. They enable the leukocytes to be caught by
the surface. Once the leukocytes are caught and begin rolling, a transition
takes place from a catch to a slip bond regime so that the latter can mediate
proper detachment from the surface.
Selectins are not only expressed by migrating leukocytes. They are
expressed early during embryonic development and enable embryos to
attach to the lining of the uterus. Failure to properly lodge in the uterus is
a prominent cause of fertilization and implantation failures. In the attachment process, carbohydrate ligands upregulated and expressed on the
surface of the uterus bind the L-selectin molecules expressed on the surface
of the embryo. The adhesion takes place under shear stresses created by
mucin secretions and, once established, sets off a chain of signaling events.
235
236
Table 10.3. Diffusible navigation markers and their receptors: The receptors are
grouped according to the ligandsnetrins, slits, semaphorins, and ephrins.
Ligand-receptor systems
Description
Netrins
DCC family
UNC-5 family
Slits
Roundabout
Semaphorins
Plexins
Neuropilins
Scatter factor receptors
Axonal repulsion
Neuronal sorting; axonal repulsion
Short range interactions; invasive growth; attraction
Ephrins
Eph receptors
to enter, or mark intermediate targets toward which the growth cones are
to turn, or provide a marker for the nal destination. The way directional
information is supplied is through concentration gradients. The basic idea,
known as the chemoafnity hypothesis, is that chemical markers are laid
down on the surfaces over which the axons navigate. These markers form
concentration gradients that are read by the growth cones to obtain the
correct heading.
237
The term short range usually refers to diffusion that spreads markers no
more than a few cell diameters from the source. The distance over which a
marker will diffuse depends on the spatial distribution of receptors. If receptors for the marker are highly expressed by nearby cells the distance traveled will be reduced over that which results from a sparse distribution of
receptors. The term contact-dependent is used to designate binding by
membrane-associated receptors on the surface of the growth cones to
membrane-associated ligands or counterreceptors expressed on the surface
of adjacent cells, and to components of the extracellular matrix such as
laminin. The distinction between a ligand and a counterreceptor is based on
whether the signaling is one-way or two-way. If it is two-way then both signaling partners are receptors. If one of the signaling partners supplies a
signal but does not transduce signals either directly or indirectly across its
own plasma membrane, then that partner is a ligand.
The modern form of the chemoafnity hypothesis has been greatly
expanded to incorporate short and long range mechanismsattraction,
repulsion, and bifunctionalityand pathnder axons, guidepost cells, and
selective fasciculation. Here is how the last process is described: Axons in
organisms such as Drosophila can grow on tracks laid down by earlier
pathnder axons. These later axons form axon bundles, or fascicles. The
axons select a particular axon bundle, bypassing inappropriate bundles in
the process, guided by molecular markers expressed on the surfaces. The
overall mechanism is thus called selective fasciculation. As the axons
approach their cellular targets they separate, or defasciculate, allowing the
axons to individually form connections.
238
Figure 10.8. Netrins, slits, and their receptors: (a) Netrins consist of a laminin-like
domain, 3 EGF domains, and a netrin C-terminus domain. (b) There are two netrin
receptors. The rst (left) is the DCC receptor contains 4 immunoglobulin (Ig)
domains followed by 6 FNIII domains. The second receptor (right) is the UNC-5
receptor. It has 2 Ig domains plus 2 thrombospondin domains. (c) Slits consist of 4
leucine-rich repeats followed by 6 EGF domains, plus an agrin-laminin-perlecan-slit
domain, plus 3 more EGF repeats and a cys knot. (d) The slit receptor Roundabout
(Robo) contains 5 Ig domains followed by 3 FNIII repeats.
239
240
241
Integrins
Boudreau NJ, and Jones PL [1999]. Extracellular matrix and integrin signaling: The
shape of things to come. Biochem. J., 339: 481488.
Giancotti FG, and Ruoslahti E [1999]. Integrin signaling. Science, 285: 10281032.
Hogg, N, et al. [2003]. T-cell integrins: More than just sticking points. J. Cell Sci., 116:
46954705.
Howe A, et al. [1998]. Integrin signaling and cell growth control. Curr. Opin Cell
Biol., 10: 220231.
Hynes RO [2002]. Integrins: Bidirectional, allosteric signaling machines. Cell, 110:
673687.
Shimaoka M, et al. [2003]. Structure of the aL I domain and its complex with
ICAM-1 reveals a shape shifting pathway for integrin regulation. Cell, 112:
99111.
Takagi J, et al. [2002]. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell, 110: 599611.
Cadherins
Boggon TJ, et al. [2002]. C-cadherin ectodomain structure and implications for cell
adhesion mechanisms. Science, 296: 13081313.
Gumbiner BM [2000]. Regulation of cadherin adhesive activity. J. Cell Biol., 148:
399403.
Yagi T, and Takeichi M [2000]. Cadherin superfamily genes: Functions, genomic
organization, and neurologic diversity. Genes Dev., 14: 11691180.
Yap AS, and Kovacs EM [2003]. Direct cadherin-activated cell signaling: A view
from the plasma membrane. J. Cell Sci., 160: 1116.
Yap AS, Niessen CM, and Gumbiner BM [1998]. The juxtamembrane region of the
cadherin cytoplasmic tail supports lateral clustering, adhesive strengthening, and
interaction with p120ctn. J. Cell Biol., 141: 779789.
242
Selectins
Kansas GS [1996]. Selectins and their ligands: Current concepts and controversies.
Blood, 88: 32593287.
Vestweber D, and Blanks JE [1999]. Mechanisms that regulate the function of
selectins and their ligands. Physiol. Rev., 79: 181213.
Problems
243
Problems
10.1 Bond dissociation strengths are not constant quantities but instead are
sensitive to the presence or absence of external forces. This dependence is captured by the expression
0
koff (F ) = koff
exp [-E (F ) kBT ].
(10.1)
244
(10.2)
(10.3)
Figure 1 for Problem 10.1. Lowering of the activation barrier by an applied force:
The applied force F acts along a direction oriented at an angle q with respect to the
reaction coordinate x, reducing the energy E by an amount Fxc os q. Because of
the dependence on the reaction coordinate x the outer barrier is driven down while
the inner barrier is barely affected. If the forces are strong enough the outer barrier
can be reduced below the height of the inner barrier, thereby revealing information
about the latters characteristics.
Problems
245
Figure 2 for Problem 10.1. Receptor-ligand bond dissociation data:The most probable unbinding forces are plotted against the logarithm of the loading rate. (a) The
data fall along a single line. (b) There is a sharp break in the data, producing two
linear regimes.
stant loading forces are applied to the bonds; that is, forces F of the
form F = rt, where r is the loading rate and t is the time. Under these
conditions the most probable unbinding force F* is related to the rate
and to the characteristic length by
F* =
kBT
ln
x
r
kBT
koff (0)
x
(10.4)
11
Signaling in the Endocrine System
248
249
naling between nerve cells. This synaptic signaling involves the diffusion of
signaling molecules (neurotransmitters) across a synaptic cleft between the
membranes of a pair of pre- and postsynaptic nerve cells. The membrane
and submembraneous regions of synapses are specialized structures highly
enriched in many different kinds of signaling molecules. Like synaptic signaling between neurons, immune system T cells use synaptic signaling when
contacting antigen-presenting cells.
250
maintains the vascular system in a plastic state. The ephrins were discussed
earlier in connection with growth cone navigation. The vascular-specic
Ephrin-B2 ligand and EphB4 receptor function in a roughly analogous
manner in angiogenesis. They are differentially expressed in endothelial
cells. The Ephrin B2 ligand functions as an arterial marker, and the EphB4
receptor operates as a venous marker.These signaling proteins help delineate
the boundary between blood vessels that become arteries and those that
become veins.
251
ture installed, and the underlying connective tissue must be remodeled and
restored.
In a skin cut, gloss keratinocytes (skin cells) migrate into the region of
injury, and upon arrival proliferate and mature. Signaling by members of
the EGF family of ligands and their receptors is essential for the proper
activities of keratinocytes during wound healing. Several EGF family
ligands are involved in repairing a skin wound. Among these are TGFa,
HB-EGF, AR, and betacellulin. These ligands are synthesized by keratinocytes, along with their EGF receptors and supply proliferation and
migratory signals, through an autocrine mechanism. In response to these
signals the keratinocytes increase their production of integrins. Binding by
these integrins to the ECM mediates substratum adherence by the keratinocytes and triggers the expression of collagesase-1.This enzyme degrades
the ECM, a step required for remodeling and repair of damaged tissue.
Continued autocrine signals maintain the expression of collagenase-1
during migration and repair.
Besides the skin, wound repair occurs in the gastronintestinal tract, in
muscle tissue following injury, and at sites of chronic inammation, to name
just a few common examples. Primary sources of epithelial growth factors
include the submaxillary salivary gland, the duodenal Brunners gland,
epithelial cells in the mammary gland, and the kidneys. Growth factors are
released into bodily uidssaliva, serum, milk, and urineand travel to
sites of injury and growth. Epithelial cells at many places in the body are
continuously renewed and growth factors are released locally near these
sites as well. In these situations the growth factors operate in autocrine and
paracrine manners rather than through an endocrine mechanism.
252
Figure 11.2. Neurotrophin receptors: The p75NTR receptor has four cysteine-rich
repeats (CR1CR4), a transmembrane segment, and a cytoplasic death domain. The
Trk receptors have a cysteine segment (C1), 3 leucine-rich repeats (LRRs), a second
cysteine segment (C2), 2 immunoglobulin-like domains (Ig1, Ig2), an insert transmembrane segment, and a cytoplasmic kinase domain. All of the neurotrophins can
bind p75NTR. NGF binds the TrkA receptor; BDNF and NT4 bind the TrkB receptor, and NT3 binds the TrkC receptor.
plasmic death domain, while the Trk receptors possess a cytoplasmic tyrosine kinase domain. The two receptors operate using different sets of
adapter proteins to convey a variety of messages into the cell as indicated
in Figure 11.3. The Trk receptors utilize Shc and Grb2 adapters, while p75
employs TRAFs and a set of four or more adapters (Figure 11.3b) to convey
death and arrest messages. In general, p75NTR receptors convey apoptotic
messages and Trk receptors convey survival messages, but as illustrated in
Figure 11.3 the full picture is far richer. These and other growth factor
receptors work together along with each other and with integrins and
cadherins to convey messages to most or all control points in the cell. These
include regulatory sites for gene expression (to promote growth and differentiation), sites of focal contact between cells and ECM (to control
movement, adhesion, cell shape, and outgrowth), and mitochondrial control
sites for apoptosis (to regulate death versus survival decisions).
253
Figure 11.3. Ligand-bound Trk and p75 neurotrophin receptors, and associated
adapters: (a) NGF-bound Trk receptor. Second messengers and associated signaling intermediates involved in activating the PKB and PKC have been omitted to
keep the gure from becoming too cluttered. (b) Neurotrophin-bound p75 receptor. Abbreviations: p75NTR-associated cell death executor (NADE), neurotrophin
receptor-interacting melanoma-associated antigen (MAGE) homolog (NRAGE),
neurotrophin receptor-interacting factor (NRIF), Schwann cell factor-1 (SC1).
extracellular region that contains one or more ligand-binding sites; a transmembrane segment; and a C-terminal cytosplasmic region that contains a
catalytic domain and several phosphorylation sites.
The RTKs that bind polypeptide growth factors are highly modular linear
arrays of domains. Among the domains present in the extracellular regions
of these glycoproteins are Ig domains, bronectin type III domains, cysteine-rich domains and the EGF ligand-binding domains. A juxtamembrane
region is situated just inside the transmembrane helix region and, as mentioned above, there is a tyrosine kinase domain near the C-terminal region.
Docking sites are opened when (auto)phosphorylation of tyrosine residues
in the activation loop of the kinase domain turns on that domains catalytic
254
255
256
257
Abbreviation
Recognition motif(s)
SH2
PTB
SH3
WW
EVH1
Phos-Tyr
Phos-Tyr, NPxY
PxxP
PPxY; PPLP; P-R; Phos-Ser
FPPPP
GYF
14-3-3
FHA
RGS
SAM
DD
PDZ
PPPPGHR
Phos-Ser
Phos-Thr
Ga
SAM
DD
PDZ, C-terminal motifs
The next-to-last entry in the table is the death domain (DD). Proteins
containing this domain convey death (apoptotis) instructions. The p75NTR
receptor shown in Figure 11.1 contains a cytoplasmic death domain (DD).
This domain enables cytoplasmic proteins bearing similar death domains
to attach via a DD-to-DD linkage. The DD is not the only module of this
sort. Instead there is an entire family of six (or seven)-helix bundle death
domains that includes the DDs, death effector domains (DEDs), caspase
recruitment domains (CARDs), and Pyrin domains (PYDs). Death
domains will be discussed further in the chapter on apoptosis.
The nal entry in the table is for the PDZ domain, named for the rst
three proteins found with this domainthe postsynaptic density protein of
95 kDa (PSD-95) found in postsynaptic terminals of neurons, the Discs
Large (DLG) protein of Drosophila, and the zona occludens 1 (ZO-1)
protein found in epithelial cells. These domains promote protein-protein
interactions through PDZ-PDZ binding and also recognize specic
sequences in the C-terminals of target proteins. Proteins containing PDZ
domains are especially prominent in synaptic terminals where they combine
with other domains to form scaffolding proteins. Members of the Shank
family of proteins serve as examples of this theme. As shown in Figure 11.4,
Shank proteins contain ve domains. Their SH3, PDZ, and proline-rich
regions either directly or indirectly bind to the three main kinds of glutamate receptors found in postsynaptic terminals, providing an anchorage
for their assembly and clustering. This assembly is anchored to the actin
cytoskeleton by intermediates that bind the ankrin repeats in the Nterminus and the proline-rich region in the middle. Finally, the SAM
258
Figure 11.4. Domain organization of a Shank protein: From the N-terminus to the
C-terminus the Shank protein has a set of ankrin repeats (Ank), an SH3 domain, a
PDZ domain, proline-rich region, and a SAM domain.
Table 11.2. Families of nonreceptor tyrosine kinases.
Family
Abl
Csk
FAK
Fes
Jak
Src
Syk
Tec
Family members
Abl, Ars
Csk, Ctk
CAKb, FAK
Fps (Fes), Fer
JAK1-3, Tyk2
Blk, Fgr, Fyn, Hck, Lck, Lyn, Src, Yes, Yrk
Syk, ZAP70
Bmx, Btk, Itk/Tsk, Tec, Txk/Rlk
domains near the C-terminus promote the linking of one Shank protein to
another through SAM-SAM binding.
259
tional modications. The Src family of NRTKs is the largest with nine
members while several other families have as few as two members. Some
NRTKs tether to the cytoplasmic face of the plasma membrane and are
covalently modied to permit attachment. Most are cytoplasmic proteins,
but Abl family members possess a nuclear localization signal (NLS) and are
found both in the cytoplasm and in the nucleus.
Figure 11.5. The Src protein in open and closed conformations: (a) The Src protein
is in an open conformation in which a crucial tyrosine residue in the kinase domain
is exposed and phosphporylated. (b) The site of tyrosine phosphorylation in the
kinase domain is blocked and the binding surfaces of the SH2 and SH3 domains are
sequestered.
260
Figure 11.6. SH2- and SH3-bearing proteins: (a) Organization of c-Src nonreceptor tyrosine kinase. (b) Organization of the Grb2 adapter protein. Proteins such as
Grb2 that lack domains that carry out enzymatic activities are referred to as
adapters. If they are very large and can bind multiple proteins they are called scaffolds. Sites where the protein can be phosphorylated on tyrosine residues are
labeled by the abbreviation p-Tyr.
261
262
Figure 11.7. Focal adhesion proteins paxillin and FAK: (a) PaxillinThe four
Lin-11, Isl-1, Mec-3 (Lim) protein-protein interaction domains in the C-terminus
mediate targeting to focal adhesions (FAs). The leucine-rich sequences, or LD
repeats, sequences of the form LDXLLXXL, where X is any amino acid residue,
found in the N-terminus domain bind FAK and proteins such as vincullin associated with the cytoskeleton. The N-terminal domain contains a number of motifs
(tyrosine phosphorylation sites and proline-rich regions) that bind to Sh2- and SH3bearing proteins. (b) Focal adhesion kinase (FAK)This protein has a focal adhesion targeting (FAT) domain in its C-terminus, and several tyrosine phosphorylation
sites and proline-rich regions in its N-terminal domain.
263
Function
Operates in the pathway that relays growth, proliferation, and differentiation
signals to the nucleus
Relays coordinating signals to the actin cytoskeleton
Shuttles mRNAs and proteins in and out of the nucleus
Regulates the targeting and docking of cargo vesicles to membranes
Regulates the formation of cargo vesicles
either GDP or GTP. More than 100 Ras superfamily GTPases have been
identied to date in eukaryotes. Each of these can be placed into one of
ve familiesRas, Rho, Ran, Rab, or Arf (Table 11.3). There are several
other groups of proteins that operate as GTPases. Chief among these are
the Ga subunits of the heterotrimeric G proteins that associate with G
protein-coupled receptors and the EF-Tu elongation factors that help
regulate protein synthesis.
Members of the Ras superfamily of GTPases carry out a variety of essential regulatory tasks. They regulate transcription, migration and focal adhesion, transport through the nuclear pore complex, assembly of the nuclear
envelope and mitotic spindle, and vesicle budding and trafcking. Ras
family members act as upstream switches in the pathway that conveys
growth and differentiation signals to the nucleus. Rho family members Rho,
Rac and Cdc42 coordinate growth and adhesion signaling. They regulate
the reorganization of the actin cytoskeleton, and coordinate its structure
with gene expression in response to extracellular signals. Ran GTPases regulate the inport and export of molecules from the cytoplasm to the nucleus.
They regulate the assembly of the nuclear envelope and also that of the
mitotic spindle. Rab proteins with over 30 family members are the largest
family of the Ras superfamily of GTPases. They are regulators of vesicle
trafcking, while Arf GTPases control vesicle budding.
264
265
Figure 11.8. GTPase cycles: (a) Switch cycle in which GTP-binding turns on the
GTPase, which can then interact with multiple effector molecules. The GTPase will
stay on until turned off by hydrolysis. (b) Assembly-controller cycle in which GTPbinding initiates interactions with an effector molecule. If the assembly is not
correct, the assembly step is aborted prior to hydrolysis, a process that is necessary
for disengagement from the substrate.
266
267
down. In bud formation, the adapter protein Bem1 is recruited to the site
of bud growth where it interacts with Ccd42 and its Cdc24 GEF. The GEF
is recruited and stabilized at the plasma membrane by the Gbg subunit activated by GPCR signaling. Both Cdc42 and Cdc24 bind to the scaffold and
a positive feedback loop is set up in which additional Bem1 proteins are
recruited. A crucial element in the choice of bud site selection is the presence of position landmarks in the cell that identify the location of the poles.
If these landmarks are absent the yeast cell is still able to form buds by a
process in which a bud site is randomly selected.
268
Figure 11.9. Ran GTPase-mediated import and export through nuclear pore complexes (NPCs): Importin (dark square) shuttles its cargo (dark circles) through the
NPC into the nucleus. In the nucleus, GTP-bound Ran GTPase triggers the release
of the cargo from the importin and shuttles the importin back out to the cytoplasm.
The GTP-bound Ran GTPase shuttles exportin (light squares) plus its cargo (light
circles) through the NPC from the nucleus to the cytoplasm where other proteins
(not shown) help dissociate the complex. The unbound exportin then diffuses back
to the nucleus.
cytoplasm. It differs from the Ras switching and Cdc42 cytoskeleton assembly in so far as its GEF and GAP actions take place in different compartments. Hydrolysis is not required for movement through the pore and
release of the cargo, but this aspect changes when Ran mediates assembly
of the nuclear envelope. In those assembly operations hydrolysis is
necessary.
269
the plasma membrane either to the lysosomes, where they are degraded, or
to endosomes, where they are recycled back to the plasma membrane. The
process of transporting plasma membrane molecules to lysosomes for
degradation is known as endocytosis, and the outward and inward movement of plasma membrane molecules is said to take place in the secretory
and endocytic pathways.
The Rab family with more than 30 known members is the largest group
of Ras superfamily GTPases. These proteins coordinate the docking and
fusion of cargo vesicles operating in the secretory and endocytic pathways.
Reciprocal pairs of proteins called v-SNAREs and t-SNAREs mediate the
fusion of cargo vesicles. The binding of the SNAREs is preceded by tethering/protection protein-binding steps that ensure that only the appropriate fusion events takes place. The Rab proteins are localized to the
cytoplasmic face of organelles and vesicles and participate in the preliminary binding operations.
The Rab protein cycle of GDP and GTP binding and release is synchronized with the movement and fusion of cargo vesicles. The combined set of
steps is illustrated in Figure 11.10. In this gure the GEFs and GAPs are
joined by a third set of accessory regulatorsGDP dissociation inhibitors,
or GDIs. These molecules bind and maintain pools of inactive Rab proteins
in the cytosol and serve as recycling chaperones. They rst help to release
the GDPases from the membranes and then shuttle them back to their site
of origin in the cytosol in preparation for their next cycle of use.
ADP-robosylation factors (Arfs) make up the fth family of Ras
GTPases. These 20-kDa proteins help regulate the formation of cargo vesicles through budding from donor membranes. In order for budding to take
place, coat proteins must be assembled over the membrane surface. These
binding agents, coat protein I (COPI), coat protein II (COPII), and clathrin,
mediate the mechanical forces that pull a membrane into a bud, and they
270
help capture membrane receptors and cargo. The Arf proteins recruit the
coat proteins to the donor membrane.
Neurotrophins
Chao MV [2003]. Neurotrophins and their receptors: A convergence point for many
signaling pathways. Nature Rev. Neurosci., 4: 299309.
Lee R, et al. [2001]. Regulation of cell survival by secreted proneurotrophins.
Science, 294: 19451948.
Zheng YZ, et al. [2000]. Cell surface Trk receptors mediate NFG-induced survival
while internalized receptors regulate NFG-induced differentiation. J. Neurosci.,
20: 56715678.
Phosphoprotein Recognition
Berg D, Holzmann C, and Reiss O [2003]. 14-3-3 proteins in the nervous system.
Nature Rev. Neurosci., 4: 752762.
Durocher D, and Jackson SP [2002]. The FHA domain. FEBS Lett., 513: 5866.
Forman-Kay JD, and Pawson T [1999]. Diversity in protein recognition by PTB
domains. Curr. Opin. Struct. Biol., 9: 690695.
Pawson T, Gish GD, and Nash P [2001]. SH2 domain, interaction modules and cellular wiring. Trends Cell Biol., 11: 504511.
Tzivion G, and Avruch J [2002]. 14-3-3 proteins: Active cofactors in cellular regulation by serine/threonine phosphorylation. J. Biol. Chem., 277: 30613064.
Yaffe MB [2002]. Phosphotyrosine-binding domains in signal transduction. Nature
Rev. Mol. Cell Biol., 3: 177186.
271
Freund C, et al. [1999]. The GYF domain is a novel structural fold that is involved
in lymphoid signaling through praline-rich sequences. Nature Struct. Biol., 6:
656660.
Kay BK, Williamson MP, and Sudol M [2000]. The importance of being proline: The
interaction of proline-rich motifs in signaling proteins with their cognate domains.
FASEB J., 14: 231241.
Macias MJ, Wiesner S, and Sudol M [2002]. WW and SH3 domains, two different
scaffolds to recognize proline-rich ligands. FEBS Lett., 513: 3037.
Mayer BJ [2001]. SH3 domains: Complexity in moderation. J. Cell Sci., 114:
12531263.
Zarrinpar A, and Lim WA [2000]. Converging on proline: The mechanism of WW
domain peptide recognition. Nature Struct. Biol., 7: 611613.
Focal Adhesions
Ilic D, Damsky CH, and Yamamoto T [1997]. Focal adhesion kinase: At the crossroads of signal transduction. J. Cell Sci., 110: 401407.
Schlaepfer DD, Hauck CR, and Sieg DJ [1999]. Signaling through focal adhesion
kinase. Prog. Biophys. Mol. Biol., 71: 435478.
Turner CE [2000]. Paxillin and focal adhesion signaling. Nature Cell Biol., 2:
E231E236.
Renshaw MW, Ren XD, and Schwartz MA [1997]. Growth factor activation of MAP
kinase requires cell adhesion. EMBO J., 16: 55925599.
272
Hancock JF [2003]. Ras proteins: Different signals from different locations. Nature
Rev. Mol. Cell Biol., 4: 373384.
Kolch W [2000]. Meaningful relationships: The regulation of the Ras/Raf/MEK/
ERK pathway by protein interactions. Biochem. J., 351: 289305.
Problems
11.1 (a) Binding interactions. One of the standard questions addressed in
investigations of signal transduction asks what interactions are
possible in a particular molecular setting. A number of proteins
Problems
273
Figure for Problem 11.1. Phosphorylated threonine and tyrosine residues are
identied by a P enclosed in a circle. The symbol PP refers to proline-rich
regions. Threonine and tyrosine phosphorylation sites with the correct anking
sequences for recognition by their kinases are indicated by the symbols p-Thr and
p-Tyr. Note that phosphorylation at the sites located in the center of the catalytic
domains of the NRTKs is required for activation.
12
Signaling in the Endocrine and
Nervous Systems Through GPCRs
276
Figure 12.1. Stereotypic representation of a rhodopsin family (Class A) G proteincoupled receptor: Shown are the seven membrane-spanning alpha helices H1H7,
three extracellular loops E1E3, and three cytoplasmic loops C1C3.
277
Calcitonin CTR
Glucagon GR
Dopamine D1 to D5
Histamine H1, H2
Melanocortin MC1, . . . , MC5
Melatonin ML1A, ML1B
Muscarinic acetylcholine
m1, . . . , m5
Neurokinen NK1, NK2, NK3
Opioid delta (d), kappa (k),
mu (m)
Prostanoid EP1, . . . , EP4,
DP, FP, IP, TP
Purine A1, A2A, A2B, A3,
P2Y1, . . . , P2Y6
Serotonin 5-HT receptor
subtypes
Somatostatin SST1, . . . , SST5
Vasopressin V1A, V1B, V2
PTH/PTHrP PTH/PTHrPR
Secretin SCTR
VIP/PACAP VIP1, VIP2
list of smaller groups with a few members each. Some of the more prominent G protein-coupled receptors belonging to the three major families are
listed Table 12.1. Most of these receptors form small subfamilies. Each
receptor has several subtypes and each subtype may be expressed in one
of a number of alternative spliced forms.
Class A contains most of the known GPCRs. It is frequently referred to
as the rhodopsin family, named for the rhodopsin molecule responsible for
transducing photons in rod cells of the retina. Receptors for a diverse spectrum of ligands including most of the amide and peptide hormones and
neuromodulators belong to this family. Class A receptors bind a number of
glycoprotein hormones such as LH, TSH, and MSH using a large extracellular domain. The glycoprotein-binding receptors differ from the GPCRs
for the smaller ligands. The latter receptors rely on extracellular loops or
utilize a binding pocket formed by the H3 to H6 helices.
Class B GPCRs, the secretin family, include calcitonin, a 32-amino acid
peptide hormone released by C cells in the thyroid gland that regulate
calcium concentrations in the blood. Secretin, a hormone released by S
cells in the small intestine, stimulates the pancreas to secrete uids that regulate acidity during digestion. Parathyroid hormone (PTH) and parathyroid hormone related protein (PTHrP) regulate bone and mineral
278
279
280
Once activated the Ga and Gbg subunits are free to diffuse laterally along
the cytoplasmic surface of the plasma membrane, and bind nearby signaling targets, or effectors. The cycle of activation and signaling is completed
with hydrolysis and reassociation of the Ga and Gbg subunits. Upon binding
Ga, the Gbg induces substantial conformational changes and increases the
afnity of Ga for GDP. When bound to Ga, the Gbg subunit cannot bind its
effectors and signal because the binding sites on Gbg for its effectors overlap
that for Ga.
Gq
G12
Gene variants
Effectors
as(S), as(L)
aolf
ai1, ai2, ai3
a0a, a0b
at1, at2
ag
az
aq, a11, a14, a15, a16
a12, a13
+AC
+AC
-AC
+PLC, +PLA2
+cGMP, PDE
+PLC
-AC
+PLC
Association
Olfactory
Brain
Retina
Gustatory
281
cyclases while others inhibit them. Still other Gbg subunits stimulate
phosopholipase C. Thus, Ga and Gbg subunits jointly determine the overall
action of a G protein on second messenger generators.
Gs
Gi
Gbg
Ca2+
I
II
III
IV
V
VI
VII
VIII
IX
+
+
+
+
+
+
+
+
+
+
-
+
-
+
+
282
Figure 12.3. Complex of the Gs alpha subunit bound to the C1 catalytic domain of
adenylyl cyclase V and the C2 domain of adenylyl cyclase II: Shown is a view of the
complex looking up from inside the cell towards the plasma membrane. The main
features of the interface between the G alpha subunit and the adenylyl cyclase
catalytic units consist of Sw I and Sw II of the Ras-like domain of G alpha that
protrude into a cleft formed by the alpha 1 to 3 helices of the C2 domain. The
gure was prepared using Protein Explorer with atomic coordinates deposited in
the Brookhaven Protein Data Bank under accession code 1AZS.
283
Figure 12.4. GRK2 and b-arrestin 2 regulators of GPCR signaling: (a) G proteincoupled receptor kinase 2 (GRK2) consists of three domains. The RGS homology
(RH) and PH domains occupy the major portions of the N-terminal and Cterminal domains, respectively. (b) b-arrestin 2 has two domains, an N-terminal
domain and a C-terminal domain. A pair of regulatory segments, R1 and R2, resides
at the ends of the N- and C-terminals. The R2 domain contains clathrin- and
AP2-binding motifs. Other binding motifs are distributed through the N- and Cterminal domains.
tion to these two domains, GRK2 (but not all GRKs) contains a C-terminal
Plectstrin homology (PH) domain. PH domains bind phospholipids and also
proteins. The GRK2 C-terminal half of this domain together with several
residues lying just outside this domain binds Gbg. The three-dimensional
crystal structure of a Gbg subunit bound to GRK2 is presented in Figure 12.5.
The structure of b-arrestin 2 is presented in Figure 12.4b. b-arrestins are
adapter proteins devoid of any catalytic ability. In place of a catalytic domain,
members of this family possess a number of protein-protein binding domains
and motifs that enable them to function as adapters and scaffolds. The barrestins contain an N-terminal domain and a C-terminal domain. Their
proline-rich region in the N-terminal domain and MAP kinase recognition
domain in the C-terminal region provide the protein with the capability of signaling to Src and MAP kinases. In addition, b-arrestins have a phosphorylation recognition domain that mediates binding to the cytoplasmic tail of the
GPCR following its phosphorylation by the GRKs. They also have clathrin
and AP2 motifs in the C-terminal region that mediate their interactions with
the molecular machinery responsible for endocytosis.
The recruitment and subsequent termination of signaling by the arrestins
is mediated by a negative feedback loop that automatically prevents excessively sustained signaling. Ligand binding activates the GPCRs, leading to
activation of G proteins. The Gbg subunits bind (Figure 12.5) and activate
284
Figure 12.5. G protein-coupled receptor kinase (GRK) bound to the Gbg subunit
of a heterotrimeric G protein: The structure of the complex determined by means
of X-ray crystallography. The three main domains of GRKthe N terminal RGS
homology (RH) domain, kinase domain and C-terminal PH domainare labeled
in the gure along with the Gb (dark gray) and Gg subunits (light gray) of the G
protein. The gure was prepared using Protein Explorer with atomic coordinates
deposited in the PDB under accession number 1OMW.
285
Figure 12.6. Receptor internalization: Activation and G protein signaling is followed by desensitization and signaling that is independent of G proteins. Ligand
binding to a GPCR activates the G protein and signaling via second messengers.
The Gbg subunit recruits a G protein-coupled receptor kinase (GRK) to the GPCR,
which is then phosphorylated by the GRK. This action creates a docking site for barrestin, which upon binding to the GPCR blocks further G protein-mediated signaling. Proteins involved in endocytosis such as clathrin and AP-2 attach to the
b-arrestin, which now acts as a scaffold for assembly for factors required for vesicle
formation and internalization. In the internalization, the ligand-receptor complexes
are dissociated and, depending on the type of GPCR, the receptors are either
degraded or recycled rapidly or slowly.
286
287
Anterior pituitary
Adrenocorticotropic hormone (ACTH)
Follicle-stimulating hormone (FSH)
Growth hormone (GH)
Lutenizing hormone (LH)
Melanocyte-stimulating hormone (MSH)
Prolactin (PRL)
Thyroid-stimulating hormone (TSH)
Endocrine pancreas
Glucagon
Insulin
Somatostatin
adipose tissue, heart muscle, skeletal muscle, brain, and lungs. The adrenal
medulla is surrounded by the adrenal cortex, a second distinct structure in
the adrenal gland. The adrenal cortex releases a large number of steroid
hormones the most important being aldosterone and cortisol that help
maintain salt balance and water homeostasis in the body.
As mentioned previously, steroid hormones do not bind to receptors on
the cell surface but instead pass through the plasma membrane and bind to
intracellular (nuclear) receptors. Peptide hormones stimulate the synthesis
and release of these hormones. Angiotensin II and III produced in the
kidneys stimulate aldosterone production, ACTH stimulates cortisol production, and the glycoprotein hormone LH stimulates the release of the
gonadotropic steroid hormones progesterone and testosterone.
Endocrine pancreas. The insulin molecule, like ACTH and its siblings, is
generated from a precursor molecule, or prohormone. The nished insulin
molecule, consisting of two chains for a total length of 51 amino acids, is
produced by cleavage from a single chain precursor, proinsulin. Once
secreted by beta cells of the Islets of Langerhans in the pancreas, insulin
binds to receptor tyrosine kinases on target fat, muscle, and red blood cells.
Glucogon, a 29-amino acid protein, is produced by alpha cells of the islets
and by cells distributed throughout the gastrointestinal tract. It binds to a
GPCR. A third hormone, somatostatin is produced by cells in the islets as
well as by cells in the hypothalamus. All three of these hormones, insulin,
glucagons, and somatostatin, inuence the ow of nutrients in the body, and
maintain glucose homeostasis by regulating the storage of glucose and its
transport in and out tissues. Insulin release is stimulated by high blood sugar
levels and triggers the uptake of glucose by its target cells. Glucagon works
in the opposite direction. Glucagon receptors are found in the liver.
288
Table 12.5. Excitatory and inhibitory neurotransmitters used in the central nervous system: Common
abbreviations for the neurotransmitters are shown in
parentheses.
Excitatory
Acetylcholine (ACh)
Glutamate (Glu)
Inhibitory
g-Aminobutyric Acid (GABA)
Glycine (Gly)
289
290
Peptides
Angiotensins
Neurokinins
Oxytocin
Substance P
Vasopressin
Nucleotides
Endogenous opioids
Adenosine
ADP, ATP
GDP, GTP
Dynorphins
Endorphins
Enkephalins
291
lar function. Production of angiotensins is stimulated by angiotensinconverting enzyme (ACE) and renin, a glycoprotein hormone produced
by the kidney and other places in the body including the brain. An infusion
of angiotensin II, the primary active angiotensin, into the brain of mammalian test subjects triggers the sensation of thirst and a craving for sodium
(salt). Laboratory animals stop what they are doing and immediately begin
drinking, and they exhibit an increased appetite for sodium when so
infused.
292
293
out the body and use a variety of G protein-coupled receptors and ion channels to sense and transduce signals.
Nociceptors are sensitive to extracellular chemical, mechanical, and
thermal environments. In response to inappropriate conditions, they transmit signals through spinal neurons to brain receptors resulting in the perception of pain. The pain pathway is the general term given to the
signaling route from the peripheral neurons located in places such as the
skin to the spinal cord to the brain. Nociceptors are distributed throughout
the body, but are generally absent in the brain, deep tissues, and visceral
organs such as the liver, spleen, and lungs. The cell bodies of the free nerves
are located in dorsal root ganglia (DRG). Signals travel from the DRG to
the dorsal horn of the spinal cord and from there to the brain. There are
two kinds of free nerve bers, Ad and C. Ad bers are thin and myelinated,
and rapidly conduct sharp pain signals. C bers are unmyelinated and
slowly conduct aching, itching, and burning signals. The C bers contain
receptors for chemical signals sent by injured cells and by cells of the
immune system. (Note: Myelinated bers are bers, or axons, that are
sheathed in myelin, a fatty protein material that insulates the axons and
promotes fast conduction of nerve pulses.)
A variety of chemical signals associated with injury and inammation are
exchanged between the nervous and immune systems. Injured cells release
some of these warning chemicals, and cells of the immune systems such as
mast cells secrete others. These messages (Table 12.7) act as local hormones
that bind to receptors on peripheral nerve cells. Some of these signaling
molecules and their receptors were discussed earlier. Those not yet examined include prostanoids, bradykinin, and purines such as adenosine. These
are discussed next.
294
Figure 12.7. Generation of fever and pain signals: Pro-inammatory signals activate phospholipase A2, which hydrolyzes membrane phospholipids (here depicted
as phospholipids in the plasma membrane) resulting in arachidonic acid.A sequence
of enzymes, beginning with COX, produces prostanoids from the arachidonic acid.
The prostanoids are short lived with half lives on the order of a minute or so and
are immediately secreted from the cell. Prostanoid receptors on peripheral nerve
endings bind the prostanoids, and the neurons convey the pain and fever signals to
the brain.
product aspirin one hundred years ago and to a host of more recent
NSAIDs such as ibuprofen and naproxen. All of the NSAIDs work the
same waythey inhibit the catalytic actions of a set of enzymes variously
called endoperoxide H synthases (PGHSs) or cyclo-oxygenases (COXs).
Two COX isoforms are targeted by anti-inammatory NSAIDs. The rst of
these, COX-1, is constitutively active and is found primarily in the stomach
and kidneys. The second, COX-2, is induced in many different cell types in
response to inammatory signals conveyed by cytokines, growth factors, and
hormones. Inhibition of COX-1 can be harmful and the aim in drug design
is to preferentially target the COX-2 isoform.
Cells release prostanoids (prostaglandins) when injured by chemical,
thermal, or mechanical agents. Prostanoids are fatty acid derivatives of
membrane lipids. As depicted in Figure 12.7, the rst step in their production is the hydrolysis of membrane lipids by phospholipase A2 (PLA2)
resulting in the release of arachidonic acid (AA). The AA intermediate is
then converted to prostanoids through the sequential actions of the COXs
and several other enzymes. The catalytic activities of PLA2 are stepped
up in response to the cytokine, growth factor, and hormonal signals. In
response the cells produce and then secrete prostanoids. During the inammatory response G protein-coupled prostanoid receptors expressed on
peripheral neurons bind prostaglandin. Signals sent on to thermoregulators
295
in the brain trigger an increase in body temperature. Aspirin and the other
NSAIDs act to lower temperature and reduce pain by inhibiting the COXs,
thereby preventing formation of and signaling by the prostanoids. The
prostanoid signals are conveyed in a paracrine fashion to their cellular
targets. For this reason prostanoids and other hormones acting in a
paracrine manner are termed local hormones.
Bradykinin is a nine-amino acid residue peptide rapidly produced
after tissue injury occurs. It is a central element in the inammatory
response. It is involved in regulating blood ow (vascular dilation), increasing vascular permeability, smooth muscle contractions, stimulating
the release of prostaglandins and other inammatory mediators, and
pain signaling. Bradykinins bind to two kinds of receptors, B1 and B2. B2
receptors are constitutive in neurons and smooth muscle cells; B1 receptors,
in contrast, are rapidly upregulated when there is tissue injury. B1 and
B2 receptors are found in free nerve endings and directly mediate pain
signaling.
Adenosine, ATP, and ADP serve as neurotransmitters and neuromodulators in the central and peripheral nervous systems. When released into
extracellular spaces these molecules bind to purine receptors. Purine receptors are divided into two families, P1 and P2, each containing a number of
subtypes (Table 12.1). Adenosine attaches to P1 receptors while ATP, ADP,
and the pyrimidines UTP and UDP bind to P2 receptors. Adenosine and
the other nucleotides carry out a large number of signaling tasks in the
body. In heart muscle, adenosine decreases heart rate and lowers blood
pressure. Adenosine is associated with sleep; during sleep deprivation
adenosine levels build up in the brain. When bound to adenosine receptors
in the brain, adenosine decreases neural activity leading to sleep. Caffeine
is structurally similar to adenosine, and is an adenosine antagonist acting
on adenosine A1 and A2A receptors. Adenosine and ATP are released into
the extracellular spaces during injury and inammation. Mast cells,
neutrophils, and free nerve endings express P1 and P2 receptors, and
adenosine helps mediate communication between the immune and nervous systems, and contributes to the pain response.
296
striking a retinal molecule in converted into helix movements, a mechanical form of energy.
The retinal molecule, unlike most other ligands, is covalently linked to
opsin and serves as a light-responsive chromophore. The retinal ligand is
buried in a pocket that lies deep in the opsin protein. When a photon strikes
the retinal chromophore it triggers a conformational change (retinal isomerization) from an 11-cis to an all-trans conguration, as shown in Figure
12.8. The retinal molecule is attached to the side chain of Lys296 situated in
the H7 helix. The movement associated with the conformational change is
appreciablewhen stabilizing the alternative all-trans conguration there
is a 4.5-movement that is reected in the positions of the cytoplasmic
portions of the transmembrane helices. In its altered conformation
rhodopsin activates the G-protein and initiates signaling in the vision
pathway. The retinal is hydrolyzed and dissociated from the opsin after
about a minute, enabling a new cycle of 11-cis-retinal attachment and
activation.
297
298
299
300
GPCR Regulation
Bockaert J, et al. [2003]. The m
agic tail of G protein-coupled receptors: An anchorage for functional protein networks. FEBS Lett., 546: 6572.
301
Grimes ML, and Miettinen HM [2003]. Receptor tyrosine kinase and G proteincoupled receptor signaling and sorting within endosomes. J. Neurochem., 84:
905918.
Hall RA, and Lefkowitz RJ [2002]. Regulation of G protein-coupled receptor signaling by scaffold proteins. Circ. Res., 91: 672680.
Hall RA, Premont RT, and Lefkowitz RJ [1999]. Heptahelical receptor signaling:
Beyond the G protein paradigm. J. Cell Biol., 145: 927932.
Koenig JA, and Edwardson JM [1997]. Endocytosis and recycling of G proteincoupled receptors. Trends Pharmacol. Sci., 18: 276287.
Lefkowitz RJ [1998]. G protein-coupled receptors III: New roles for receptor
kinases and arrestins in receptor signaling and desensitization. J. Biol. Chem., 273:
1867718680.
Lodowski DT, et al. [2003]. Keeping G proteins at bay: A complex between G
protein-coupled receptor kinase 2 and Gbg. Science 300: 12561262.
Luttrell LM, and Lefkowitz RJ [2002]. The role of b-arrestins in the termination and
transduction of G protein-coupled receptor signals. J. Cell Sci., 115: 455465.
Phototransduction
Baylor D [1996]. How photons start vision. Proc. Natl. Acad. Sci. USA, 93: 560565.
Clapham DE, Runnels LW, and Str
bing C [2001]. The Trp ion channel family.
Nature Rev. Neurosci., 2: 387396.
Hardie RC, and Raghu P [2001]. Visual transduction in Drosophila. Nature, 413:
186193.
302
Tastants
Adler E, et al. [2000]. A novel family of mammalian taste receptors. Cell, 100:
693702.
Chandrashekar J, et al. [2000]. T2Rs function as bitter taste receptors. Cell, 100:
703711.
Chaudhari N, Landin AM, and Roper SD [2000]. A metabotropic glutamate receptor variant functions as a taste receptor. Nature Neurosci., 3: 113119.
Lindemann B [2001]. Receptors and transduction in taste. Nature, 413: 219225.
Margolskee RF [2002]. Molecular mechanisms of bitter and sweet taste transduction. J. Biol. Chem., 277: 14.
Nelson G, et al. [2001]. Mammalian sweet taste receptors. Cell, 106: 381390.
Problems
12.1 The b-arrestins can function as switches that rst shut down signaling
through the G proteins and then turn on signaling through growth
pathways. When switching to non-G protein modes of signaling, the
arrestin molecule and the GPCR serve as a platform for the assembly
of Src and other signaling proteins. The essential features that mediate
these associations are the presence of peptide sequences that bind to
SH2, SH3, and PDZ domains, either in the third intracellular loop or
in the cytoplasmic COOH-terminal tail, and the Src non-receptor tyrosine kinase that can bind to the proline rich motifs of the b-arrestins
by means of its SH3 domain. Construct a growth pathway leading to
the nucleus that begins at the GPCR and is routed through Src that
binds to the b-arrestins.
12.2 One of the G protein alpha subunits, Gs, stimulates adenylyl cyclases
to produce cAMP, which activates protein kinase A. In b2 adrenergic
GPCR (b2AR) signaling, protein kinase A phosphorylates the GPCR,
and this negative feedback loop not only desensitizes the receptor but
also switches its G protein preference from Gs to Gi. What are two,
Problems
303
13
Cell Fate and Polarity
There are approximately 1014 cells in the human body. Some of these are
heart cells; others are liver, kidney, nerve, or muscle cells. During development sets of identical progenitor cells undergo different cells fates. They
diverge at the correct time to form different cell types along the appropriate body axes and boundaries. Mutual signaling between these cells drives
many of these decisions so that each cell knows what kind of cell to become.
These signals are transmitted from cell to cell, transduced across the plasma
membrane, and sent on to the nucleus where they activate specic sets of
developmental genes. Some genes are turned on and others are turned off
in response to these signals.
A small number of signaling pathways guide embryonic development. In
the rst part of this chapter, four pathways of particular importance with
respect to developmentNotch, transforming growth factor-b (TGF-b),
Wnt, and hedgehogwill be examined. These pathways regulate the programs of gene expression so that at the correct time in the right place cells with
the same propensity for a particular cell fate give rise to daughters exhibiting
differences in morphology and the mix of proteins being expressed.
A variety of stratagems are used to achieve the developmental goals. One
of these is to lay down gradients of signaling proteins either on cell surfaces
or in extracellular spaces that help determine cell fate when they activate
receptors on a cell. These signaling proteins are known as morphogens.
Another stratagem is to utilize hierarchical sequences of gene expression
so that over time different progeny will become different kinds of cells. The
focus in the rst part of the chapter will be on how signals are sent from
the cell surface to the nucleus through the four pathways. In the second part
of the chapter, the goal will be to see how morphogen gradients and hierarchical patterns of gene expression guide cell fate decisions.
The Notch, TGF-b, Wnt, and Hedgehog signaling pathways are named
either for the transmembrane receptor or for the molecules that serve
as ligands for the receptors. These four pathways are highly conserved in
multicellular organisms. They have been studied extensively in the y
(Drosophila), worms (C. elegans), and vertebrates. A variety of rather color305
306
ful names have been given to signaling proteins belonging to these pathways.
The names are usually derived from the types of developmental defects seen
in Drosophila when the genes encoding the proteins suffer a mutation, usually of the loss-of-function type. For example, when the Hedgehog gene is
mutated, a spiky process called denticles is seen, and hence the name hedgehog was given to the protein. In the Notch pathway, partial loss-of-function
defects in the Notch receptor produced notches in the wing. Defects in the
Groucho protein, a downstream-acting element that participates in several
pathways, result in the production of bristles around the eye. These aberrant
structures resemble the eyebrows of the well-known comedian and hence the
name Groucho was given to the gene and its protein product.
Vertebrates
Drosophila
C. elegans
Delta1, 2, Jagged1, 2
Notch14
CBF-1
Delta, Serrate
Notch, LIN-12
Su(H)
LAG-2, Apx-1
GLP-1
LAG-1
307
308
Figure 13.2. Signaling through the Notch pathway: The sending cell expresses the
Notch ligand Delta or Jagged, while the receiving cell expresses the Notch receptor. The extracellular chain of Notch binds the ligand and in response the transmembrane segment is cleaved at several places to form the NICD. The NICD,
together with Su(H), translocates to the nucleus where they interact with several
other proteins to activate gene transcription. As depicted in the gure, they may
promote gene transcription by displacing corepressors.
309
Figure 13.3. Lateral inhibition through cell-to-cell signaling and positive feedback:
(a) Because of stochastic uctuations there are more Notch receptors on the B cell
than on the A cell and, other things being equal (i.e., the density of Delta ligands
on the two cells being the same), signaling into the B cell is stronger than in the A
cell. (b) In the B cell, Notch signal transduction upregulates Notch receptor expression and reduces Delta ligand expression. On the A cell the strength of Notch signaling fails to compensate for the decay of Notch receptors over time. Thus on the
B cell Delta ligand expression goes down while on the A cell Notch receptor expression declines. (c) This tendency is amplied over time leading to an A cell expressing Delta ligands and a B cell expressing the Notch receptors.
310
ment in cleaving the amyloid precursor protein (APP) leading to the release
of the Ab amyloid protein, an important step in the onset of Alzheimers
disease. Like the Notch process depicted in Figure 13.1 the APP undergoes
several cleavages. These are mediated by enzymatic complexes referred to
as a-secretases, b-secretases, and g-secretases. The Presenilins are part of
the g-secretase complex, which has three other membersNicastrin, Aph1 and Pen-2. As illustrated in Figure 13.4 the Presenilins are 8-pass transmembrane (TM) proteins proteolytically cleaved into two chains. A pair of
aspartate residues positioned in opposition to one another is crucial for Presenilins g-secretase actions. Notch and APP pass through the gap formed
by the TM segments containing the aspartates, and are cleaved. The Presenilins carry out their catalytic activities together with the single-pass Nicrastrin protein, the 7-pass Aph-1 protein, and the 2-pass Pen-2 protein, which
help to assemble and stabilize the complex.
The APP is cleaved twice, one in its extracellular domain just outside the
membrane and the other near the middle of the intermembrane region. In
the rst step, the APP proteins are cleaved at one of two alternative extracellular locations termed the a- and b-sites. The g-secretases are responsible for subsequent cleavages within the transmembrane segment. In the
case of cleavage at b-sites, but not a-sites, the products are 40- and 42-amino
acid residue forms of the Ab amyloid protein. The 42-residue form, favored
by the mutated Presenilins, is especially prone to form clumps due to
exposure of hydrophobic patches, as was discussed in Chapter 5. The asecretases and b-secretases responsible for cleaving the APPs at the aand b-sites are members of two other families of proteases. One or more
members of the a disintegrin and metalloprotease (ADAM) family are
the a-secretases, while aspartyl proteases belonging to the membraneassociated aspartyl protease (memapsin) family serve as the b-secretases.
The ADAM enzymes are members of a large group of multidomain
enzymes that possess not only metalloproteinase domains, but also integrinbinding regions and a cytoplasmic domain that bind a variety of signaling
311
312
Figure 13.5. Signaling through the TGF-b pathway: Binding of a ligand (TGF-b)
dimer to a TbRII-TbRI receptor complex leads to phosphorylation of serine/threonine residues in the GS region of the TbRIs resulting in the activation of the kinase
domain. The activated receptors phosphorylate the Smad3 proteins leading to the
latters dissocation from the SARA anchors. They form a heterotrimeric complex
with Smad4, and in that form translocate to the nucleus where they form a scaffold
for assembly of transcriptional activators and cofactors such as CPB/p300 and Ski
to induce the transcription of target genes.
313
number of different adapter proteins. One of these, the Smad anchor for
receptor activation (SARA), couples Smad2/3 to the activated receptor
complex. The SARA protein possesses a lipid-binding FYVE domain that
enables it to tether to the plasma membrane. Two other domains permit it
to bind simultaneously to the receptor and to the Smad protein. Once the
Smad proteins have been recruited to the receptor complex they are phosphorylated, triggering their release into the cytosol (Figure 13.5).
Table 13.2. The TGF-b signaling pathway: As indicated in the table, the TGF-b
pathway branches at the Type I receptors (Type I Rs) level into an Activin-like
branch and a BMP-like branch. AbbreviationsActivin receptor-like kinase
(ALK); bone morphogenetic protein (BMP); transforming growth factor (TGF);
receptor (R); inhibitory (I).
Activin
TGF-b
TGF-b
BMP
Ligands
Activins
TGF-bs
TGF-bs
BMPs
Type II Rs
ActII/IIB
TbRII
TbRII
BMPRII,
ActRII/IIB,
MISRII
Type I Rs
ALK4
ALK5
ALK1
ALK3, ALK6,
ALK2
R-Smad
Smad2, Smad3
Smad2, Smad3
Smad1, Smad5,
Smad8
Smad1, Smad5,
Smad8
Co-Smad
Smad4
Smad4
Smad4
Smad4
I-Smad
Smad7
Smad7
Smad6, Smad7
Smad6, Smad7
314
315
Figure 13.6. Wnt signal receptors and transducers: (a) LRP coreceptor. (b) Frizzled receptor. (c) Strabismus co-receptor. The rectangle attached to the cytoplasmic
C-terminal tail of the receptor denotes a PDZ binding domain. (d) Dishevelled
adapter. AbbreviationsDishevelled, Egl-10, and Pleckstrin (DEP); Dishevelled
and axin (DIX). The DIX domain mediates attachment to the cytoskeleton, and the
DEP domain promotes binding to the plasma membrane.
316
Figure 13.7. Signaling through the Wnt b-catenin and PCP pathways: Ligand
binding activates either the canonical b-catenin pathway or the planar cell polarity
pathway, depending on the actions at the Dishevelled switching point. In the canonical pathway, Dishevelled acts through the CKI kinase to inuence the decision
whether LEF/TCF gene transcription is repressed or activated. Alternatively,
Dishevelled acts through the small GTPase protein RhoA and downstream MAP
kinases such as JNK to promote cytoskeleton polarization and gene expression.
axin, the adenomatous polyposis coli (APC) protein, and two serine/threonine kinasesGSK3 and CKI. This module works in the following way. The
kinases phosphorylate the other members of the complex. Phosphorylation
of axin stabilizes it; phosphorylation of APC enhances its interactions with
b-catenin. b-catenin cannot signal because phosphorylation tags it for proteolytic destruction. As a consequence of the phosphorylations, b-catenin
does not translocate to the nucleus and stimulate transcription. Dishevelled
acting through CKI destabilizes the complex, and leads to dephosphorylation of b-catenin. The number of mobile and untagged b-catenin molecules
increases, and they are able to translocate to the nucleus and stimulate transcription of TCF/LEF genes (Figure 13.7).
The presence of two serine/threonine kinases in the complex leads to
a two-step phosphorylation/degradation process. CKI acts rst to phosphorylate substrate residues within the complex. This action primes the
system for subsequent phosphorylation by GSK3, which is then followed
by the recruitment of proteolytic elements to b-catenin. In more detail, CKI
phosphorylates b-catenin at Ser45. This action enables GSK3 to phosphorylate b-catenin at three neighboring sites (Ser33/Ser37/Thr41). In the
absence of the priming CKI phosphorylation, GSK3 cannot phosphorylate
b-catenin at the aforementioned sites.
317
318
Figure 13.8. Hedgehog receptors Patched and Smoothened: (a) Patched receptor.
(b) Smoothened receptor in a closed conformation. (c) Smoothened receptor in an
open conguration.
events. For example, the Hh pathway is responsible for body, wing, eye,
genitals, and leg patterning in Drosophila while the Shh pathway regulates
eye, portions of the brain, hair, lung, gut, bladder, and urethra patterning in
mammals.
The Patched receptor is a 12-pass transmembrane protein (Figure 13.8).
It functions together with a partner receptor called Smoothened. The
Smoothened protein is a 7-pass transmembrane receptor similar to the
Wnt receptor Frizzled. Patched is responsible for ligand binding and
Smoothened is responsible for transducing the signal across the plasma
membrane. Two different conformations of the Smoothened receptor are
depicted in Figure 13.8. In the rst, (b), the cytoplasmic tail cannot bind to
the downstream signaling partners, but, in the second, (c), binding sites
along the cytoplasmic tail become available. In the absence of ligand
binding, Patched acts catalytically to maintain Smoothened in its closed
conformation, but ceases to do so when bound to Hedgehog.
The cytoplasmic tail (CT) of Smoothened in its open conformation interacts with two cytoplasmic proteinsCostal-2 (Cos2) and Fused (Fu). The
rst of these, Cos2, is a kinesin-like protein that binds to microtubules while
the other, Fu, is a serine/threonine kinase. The Cos2 protein functions as a
scaffold for assembly of several signaling elements. These include, besides
Smoothened, CT and Fused, Suppressor of Fused (Su(Fu)) and a downstream-acting transcription factor called Cubitus interruptus (Ci). Two
additional serine/threonine kinasesPKA and GSK3complete the
specication of this Hedgehog signaling node (Figure 13.9).
319
Figure 13.9. Signaling through the Hedgehog pathway: In the absence of ligand
binding, Patched (Ptc) represses Smoothened (Smo), depicted in the gure as acting
through a small molecular (Sm mol) intermediary. In response to ligand binding,
the repression is relieved and Smo undergoes a conformational change from a
closed conguration to an open one that can bind Cos2 and Fu. This binding action
prevents phosphorylation of Ci by PKA and GSK3. The complex dissociates and
the full length 155 kDa CiA translocates to the nucleus where it promotes transcription. In the absence of ligand binding, the Ci protein is cleaved following phosphorylation, and the 75 kDa CiR protein translocates to the nucleus where it
represses transcription.
Figure 13.10. Organization of the Drosophila Cubitus interruptus (Ci) protein: Five
tandem zinc ngers situated in the N-terminal half of the protein are responsible
for binding DNA. A CBP binding domain located in the C-terminal portion of Ci
binds a CBP cofactor. Cleavage of the protein produces the CiR protein containing
the N-terminal portion and acting as a transcriptional repressor. Three of the many
S/T phosphorylation sites (pSer) are shown. The sites indicated are the priming sites
phosphorylated by protein kinase A (PKA). Sites located both N-terminal and
C-terminal to these sites are phosphorylated by GSK3.
320
with Cos2, Fu, and Su(Fu) that is bound to microtubules. In the absence of
Hh signaling, another member of the signaling pathway, protein kinase A
(PKA) phosphorylates Ci. This phosphorylation event tags Ci for proteolysis. The result of this process is the formation of a 75-kDa CiR protein
(Figure 13.9), which translocates to the nucleus where it functions as a
repressor of the transcription of several genes. When Hh is present, phosphorylation of Ci by PKA is blocked, the complex dissociates, and the fulllength 155-kDa protein is left intact and free to move into the nucleus. This
intact CiA protein functions as an activator of transcription of a number of
genes including patched, dpp, and wg.
321
Function
Members
Maternal effect
Gap
Pair rule
Segment
polarity
Hox cluster
322
323
Figure 13.11. Schematic depiction of a Drosophila oocyte: Shown in the gure are
the polynuclear nurse cells, oocyte, and posterior array of follicular cells. Bicoid
mRNAs are laid down in a strip in the anterior end and diffuse towards the posterior pole to form a concentration gradient. Oskar mRNAs and proteins are laid
down at the posterior pole and diffuse out to form a countergradient.
324
325
body of the developing larva. As is the case for the other primary pair rule
genes, the regulatory region for even-skipped (eve) contains a set of special
binding sites, called enhancer elements, for transcription factors, one for
each stripe. These enhancers correspond in a unique way to the gradient
information in the region of the stripes.
Gap and maternal effect genes both regulate the pair rule gene expression patterns.The gene regulatory region (enhancer) for eve in stripe 2 illustrates the general theme. Bicoid and Hunchback bind to sites in the
enhancer and function as activators of gene expression. Giant and Kr
ppel
also bind to sites in the enhancer and operate as repressors of gene expression. Since Giant is expressed at the anterior end and Kr
ppel in the middle,
stripe 2 is inhibited in those regions but is encouraged by Bicoid and
Hunchback in a stripe in the anterior region located in between the two
repressed regions. Similarly, enhancers of the other stripes reect the gradients of the corresponding sets of maternal effect and gap genes in the
regions where the other stripes are to be sited. The net result is the emergence of seven stripes of expression for eve, and similarly, seven stripes for
hairy and seven stripes for runt. The gene products appear in regions that
partially overlap one another and so they operate in combinatorial manner
to delineate the parasegments.
326
327
fully timed. The timing activity keeps the movement of the wave in phase
with the time needed for development of the somites.
328
Kalderon D [2002]. Similarities between the Hedgehog and Wnt signaling pathways.
Trends Cell Biol., 12: 523531.
Ma D, et al. [2003]. Fidelity in planar cell polarity signaling. Nature, 421: 543547.
Mlodzik M [1999]. Planar polarity in the Drosophila eye: A multifaceted view of
signaling specicity and cross-talk. EMBO J., 18: 68736879.
Moon RT, Brown JD, and Torres M [1997]. Wnts modulate cell fate and behavior
during vertebrate development. Trends Genet., 13: 157162.
Wallingford JB, Fraser SE, and Harland RM [2002]. Convergent extension: The
molecular control of polarized cell movement during embryonic development.
Dev. Cell, 2: 695706.
Hedgehog Signaling
Collins RT,and Cohen SM [2003].The secret life of Smoothened.Dev.Cell,5:823824.
Hammerschmidt M, Brook A, and McMahon AP [1997]. The world according to
Hedgehog. Trends Genet., 13: 1421.
Ingham PW [1998]. Transducing Hedgehog: The story so far. EMBO J., 17:
35053511.
Ingham PW, and McMahon AP [2001]. Hedgehog signaling in animal development:
Paradigms and principles. Genes Dev., 15: 30593087.
Taipale J, et al. [2002]. Patched acts catalytically to suppress the activity of
Smoothened. Nature, 418: 892897.
Morphogens
Lawrence PA, and Struhl G [1996]. Morphogens, compartments, and pattern:
Lessons from Drosophila? Cell, 85: 951961.
Teleman AA, Strigini M, and Cohen SM [2001]. Shaping morphogen gradients. Cell,
105: 559562.
Problems
329
Guss KA, et al. [2001]. Control of a genetic regulatory network by a selector gene.
Science, 292: 11641167.
Hirata H, et al. [2002]. Oscillatory expression of the bHLH factor Hes1 regulated
by a negative feedback loop. Science, 298: 840843.
Saga Y, and Takeda H [2001]. The making of the somite: Molecular events in vertebrate segmentation. Nature Rev. Genet., 2: 835845.
Problems
13.1 The core components and basic features of the four primary developmental pathways have now been introduced. The underlying picture
is a fairly simple one in which ligand binding at the plasma membrane
triggers a sequence of signaling events culminating in the activation
of gene transcription in the nucleus. The signaling pathways possess a
number of characteristics that enable them to guide the development
of complex metazoans possessing a large variety of different cell types
organized into tissues and organs. The rst of these characteristics is
that signaling is context dependent. The term context refers to the
mix of proteins being expressed. How a cell responds to a signal
depends on what other signaling events are taking place at the same
time and in previous times that have altered the mix of proteins being
expressed. How might context dependence come into play at the
plasma membrane top inuence signaling?
13.2 Control points are situated at the plasma membrane, in the cytoplasm,
and in the nucleus. Examples of cytoplasmic control points are the
beta catenin complex formed in the Wnt signaling pathway and the Ci
signaling complex formed in the Hedgehog signaling pathway. These
along with MAP kinase and NF-kB modules function as major signaling nodes. Give some examples of how cellular context may inuence what happens at these nodes in the signaling pathway.
13.3 Two kinds of posttranslational modicationsphosphorylation and
proteolysis are especially prominent in the four developmental pathways. These modications operate individually and jointly in these
pathways. List the various ways these modications inuence signaling.
13.4 Despite the presence of different proteins, the developmental pathways have many features in common. In response to ligand binding, a
series of signaling events takes place resulting in the translocation
from the cytoplasm to the nucleus of a transcriptional factor. In these
pathways downstream signaling elements can often function both as
activators of transcription or as repressors, with the choice depending
on cellular context. How does context come into play in the nucleus?
How was this dual capability used in the pathways discussed in this
chapter?
14
Cancer
Cancers arise from malfunctions in the cell control layer that lead to the
unregulated proliferation of cells. The underlying causes of cancers are
mutations and other alterations in DNA, and attendant inappropriate
expression levels, of genes encoding proteins that either promote growth or
restrain it, or direct the apoptosis machinery, or are responsible for DNA
damage repair and signaling, and chromatin remodeling.The mutations may
be heritable, that is, they may be present in the germ cells, or they may be
produced in somatic cells.
DNA can be damaged in several ways. Hydrolytic processes and oxidative byproducts of normal cellular metabolism can damage DNA. Ionizing
radiation from cosmic rays and from natural-occurring radioactive materials in the soil, water, and air such as uranium, thorium, and radon can
damage DNA. In addition, ultraviolet (UV) radiation from the sun can
damage DNA. The main step leading to DNA damage by environmental
and endogenous stimuli is the generation of oxidative free radicals near the
DNA. Free radicals are molecular species with unpaired electrons, making
them highly reactive. The most damaging of these reactive oxidative species
(ROS) is the hydroxyl radical. When produced in the vicinity of a DNA
molecule the hydroxyl radicals and other ROS attack sugars and bases producing single strand breaks, base losses, and modied bases. In addition to
this ROS mechanism, ionizing radiation such as X-rays and gamma irradiation can directly generate double strand breaks in DNA. When a cell that
has been damaged divides without having the damage repaired the changes
are carried on to its daughter cells and become a mutation.
In metastasis, malignant cancer cells break away from where they are
immobilized, enter the circulatory system, and invade other organs. They
form colonies, or secondary tumors, at the new locations and cause damage
to their neighbors by appropriating their sources of nutrition. Cancers
derived from epithelial cells are the most common type of cancer. In order
for the cancerous epithelial cells to break away from their initial location
they must detach from other epithelial cells and from the ECM. To do so
they must disrupt the cell-to-cell adhesive contact established by E331
332
14. Cancer
Figure 14.1. Matrix metalloproteinase structure: Most MMPs contain an Nterminal signal peptide (not shown) that targets the enzyme for secretion, followed
by a prodomain. The catalytic domain contains a zinc-binding domain and all MMPs
except MMP7 and MMP26 contain a Hemopexin domain C-terminal to the catalytic
domain and separated from it by a linker region. Dashed boxes represent domains
present in some MMPs but not others. Included in this set are Furin-like domains,
Type II Fibronectin repeats, and transmembrane + cytoplasmic segments.
333
Function/pathway
Class
Cancer role
Growth
Growth
Growth
Growth
Growth
Growth
Apoptosis
Apoptosis
Repair
Repair
Repair
Repair
Repair
Controller
Controller
o
o
o
ts
o
o
ts
o
c
c
c
c
c
ts
ts
Many cancers
Sarcomas
Leukemias
Colorectal cancers
Colorectal cancers
Many cancers
Many cancers
Many cancers
Colon cancers
Colon cancers
Ataxia telangiectasia
Nijmegen breakage syndrome
Breast/ovarian cancers
Most cancers
Most cancers
334
14. Cancer
335
Figure 14.2. Structure of Ras in a complex with its Sos GEF as determined using
X-ray crystallography: Ras is shown in light gray while Sos is depicted in black. Small
lled circles and squares superimposed on the gure serve to outline Sw 1 and Sw
2, respectively. The gure was generated using Protein Explorer with atomic coordinates deposited in the Brookhaven Protein Data Bank (PDB) under accession
code 1BKD.
Figure 14.3. Structure of Ras in a complex with its GAP as determined using Xray crystallography: Ras is shown in light gray while the RasGAP is depicted in
black. Small lled circles and squares superimposed on the gure serve to outline
Sw 1 and Sw 2, respectively. The Gly12 residue is located at the tip of the Ras Ploop while a second crucial residue, Glu61, located in the Ras Sw 2 region in close
opposition to the arginine nger loop of the RasGAP. The gure was generated
using Protein Explorer with atomic coordinates deposited in the Brookhaven PDB
under accession code 1WQ1.
336
14. Cancer
molecule is plentiful and binds in the pocket that was vacated by the GDP
molecule.
Mutations of the glycine residue at position 12 in the Ras chain convert Ras
into a form that is active all the time.That is, the RasGAP is unable to turn off
Ras.The reason for this can be discerned from the crystal structure exhibited
in Figure 14.3. The Gly12 residue is positioned at a crucial place at the very
end of the P loop.Because of its small size,replacement of glycine by any other
residue blocks the arginine in the nger from interacting with the ATP
molecule bound in the cleft, and hydrolysis is consequently impeded.
337
In order for a solid tumor to grow and thrive it must have an adequate
blood supply. In response to this need for vascular expansion, tumor angiogenesis takes place. The expression of messenger RNAs for VEGF ligands
is enhanced in most human tumor cells. Increased VEGF mRNAs are
present in rapidly growing glioblastoma multiform brain tumors, and in
cancers of the lung, breast, gastrointestinal tract, female reproductive
organs, thyroid gland, and urinary tract.
Growth factor receptor dimerization is a critical step in relaying signals
conveyed by polypeptide growth factors into the cell interior.As discussed in
Chapter 11, receptor tyrosine kinases are brought into close physical proximity through ligand binding, resulting in the formation of receptor dimers or
oligomers. Autophosphorylation in the activation loop occurs next followed
by recruitment of cytoplasmic signaling molecules. In the absence of a ligand
the receptors do not dimerize and there is no signaling across the plasma
membrane. In contrast to this normal situation, spontaneous dimerization
occurs in many cancers. In these abnormal situations the receptors dimerize
in the absence of ligand binding. Ligand-free dimerization can be produced
in several different ways. Most often it is generated through overexpression
of receptors arising as a consequence of gene amplication.It can also be generated through point mutations and exon deletions.
Epidermal growth factor receptors (EGFRs) undergo spontaneous
dimerization in many cancers including those of the breast, lung and ovarian
cancers and gliomas, and brain tumors of glial origin. Amplication of the
EGFR (ErbB1) gene occurs in about 40% of gliomas, and the amplication of the ErbB2 gene takes place in about 30% of breast cancers. In many
of the brain tumors, gene rearrangements accompany gene amplication
and these alterations often involving truncations of portions of the molecule. The main effect of spontaneous dimerization is to activate a pathway
that sends inappropriate growth/proliferation signals to the nucleus.
338
14. Cancer
tivity between adhesion and growth factor receptors are a6b1 and a6b4 and
EGFRs, NCAM, and N-cadherins with FGFRs, and VE-cadherins with
VEGF receptors. One result of this form of association is the ability of integrins and/or growth factor receptors to convey signals into the cell without
having to engage their natural ligands. Clustering brings the receptors into
close proximity with one another, and promotes phosphorylation and the
recruitment of cytoplasmic signaling transducers, thereby alleviating the
need for ligand engagement.
A second consequence of the growth factor receptoradhesion molecule
clustering is the strengthening of signals that would otherwise be too weak
to elicit a cellular response if conveyed by one or the other alone. An
example of this form of cooperativity is that which occurs between Met and
a6b4 integrins. Recall from Chapter 10 that Met is the receptor for HGF/SF,
a set of diffusible ligands that are central participants in invasive growth
and branching morphogenesis. SF does not stimulate growth, but rather
triggers the dissociation, or scattering, of cells. By forming clusters the cytoplasmic segments of the Met receptors and integrins come into close
contact; they are able to promote phosphorylation and provide multiple
docking sites for adapters and other cytoplasmic signaling elements. In this
second signaling role, the a6b4 acts as an amplier to increase the magnitude of the cellular response to a growth signal and promote invasive
growth independent of binding to the ECM.
339
340
14. Cancer
341
System(s)
BER
NER
Mismatch repair
MMR
HR, NHEJ
Description
Removes bases damaged by UV, X-rays,
oxygen radicals, and alkylating agents
Removes DNA lesions brought on by
environmental agents
Repairs damage occurring during DNA
replication and meiotic recombination
Repairs double-strand breaks caused by
ionizing radiation, oxidative stresses, and
other environmental factors
bulky DNA lesions and damage arising from environmental agents such as
polycyclic aromatic hydrocarbons compounds contained in cigarette
smoke. Portions of chromosomes bearing genes that are actively undergoing transcription receive greater NER attention than DNA segments containing genes that are only rarely transcribed. Several disorders including
skin cancers and Cockaynes syndrome are associated with mutations in
genes encoding members of the NER system.
The mismatch repair system monitors and treats damage occurring
during DNA replication and meiotic recombination. Among AGTC combinations corrects A-G and T-C mismatches, as well as improper sequence
insertions and deletions. Mutations in this repair systems lead to increases
in the mutation rate and to cancer development. Mutations in two members
of the MMR system, hMLH1 and hMSH2, are prominently linked to colorectal and other cancers.
The machinery for repairing double strand breaks is utilized for several
purposes in the cell. It is central to V(D)J recombination in lymphocytes.
DNA undergoing replication is subject to breaks and the machinery for
repairing DNA participates in remedying broken replication forks. This
machinery is involved in genetic recombination, the process whereby segments of DNA are exchanged between chromosomes. It is involved in both
mitotic recombination, involving sister chromatids, and meiotic recombination, involving homologous chromosomes. Homologous recombination and
nonhomologous end joining are responsible for repairing DNA doublestrand breaks. HR utilizes regions of DNA sequence homology from the
sister chromatid to repair damaged DNA in an error-free manner. NHEJ
does not utilize extensive regions of sequence homology to effect repairs
and is not necessarily error-free.
Double-strand breaks (DSBs) can be generated endogeneously by oxidative agents and environmentally by ionizing radiation. This form of damage,
while not as common as the other forms of DNA injury, can be extremely
harmful. DSBs can lead to chromosomal translocations, deletions, and fragmentation. Mutations in several members of the machinery that repair
342
14. Cancer
Function
ATM/ATR
Signaling
Rad50
HR, NHEJ
Mre11
NBS1
Rad51
HR, NHEJ
HR, NHEJ
HR
Rad52
Rad54
BRCA1,2
Ku70
HR
HR
HR
NHEJ
Ku80
DNA-PKcs
XRCC4
DNA ligase IV
NHEJ
NHEJ
NHEJ
NHEJ
Description
Phosphorylates p53, NBS1, and BRCA1 in
response to DSBs
Forms an end-processing complex with Mre11 and
NBS1
Endonuclease activity
Regulatory
Searches for DNA homology; forms a complex
with Rad52, Rad 54 and BRCA1,2
Stimulates Rad51 activity
Chromatin remodeling
Regulatory; chromatin remodeling
Forms a heterodimer with Ku80; recruits and
activates DNA-PKcs
KU70 and KU80 bind to the free ends of the DSB
Regulatory; signals p53
Forms a complex with DNA ligase IV
Ends reattachment
343
with regulatory functions and others with repair functions. The modules
listed in Table 14.3 work synergistically and sequentially to make the repairs
to the DNA. The early steps in operation of this repair system are depicted
in Figure 14.4.
The rst module, referred to as the Mre11 complex, is composed of the
Rad50, Mre11, and NBS1 proteins. This module participates in both homologous recombination and nonhomologous end joining along with several
other DNA strand manipulations involved in mitosis and meiosis. The key
function of this complex is to form exible bridges between ends of broken
DNA strands. The Rad50 proteins contain hooks, which join opposing
Rad50 proteins protruding from the ends of broken DNA strands. The
hooks join pairs of Rad50 proteins in the middle of the break while the
ends of the Rad50s remain attached to the DNA strands via the Mre11
proteins.
The next module is the Rad52 group. The Rad52 family of proteins is
responsible for homologous recombination. The rst step in HR is the
coating of the single strand DNA (ssDNA) with molecules of replication
protein A (RPA). In the next step, molecules of Rad51 and BRCA2 are
loaded onto the strands so that Rad51 displaces RPA. Both RPA and Rad51
remove secondary structure from the DNA to facilitate the pairing of sister
344
14. Cancer
345
Figure 14.5. Crystal structure of the COOH terminal portion of the BRCA2 tumor
suppressor protein: Shown are the main secondary structure elements helices
(corkscrew-shaped ribbons) and strands (at arrow-shaped ribbons), and the overall
organization of the COOH terminal portion of the molecule into ve domains. The
tower domain binds DNA and is the locus of a large number of cancer-inducing
mutations. The oligonucleotide/oligosaccharide binding (OB) domains bind ssDNA.
Two fragments of a DSS1 protein found in association with BRCA2, and needed
for crystallization, are included in the gure. The gure was generated using Protein
Explorer. The Brookhaven PDB accession number is 1MIU.
346
14. Cancer
rst two letters of a patient found to be suffering from an autoimmune disorder associated with the Ku proteins.
DNA-PKcs and ATM are structurally similar, and both molecules are
capable of binding DNA. DNA-PK not only participates in repair of DSBs
caused by ionizing radiation but also is involved in V(D)J recombination
used to generate diversity in cells of the immune system. A mutation in
the kinase domain produces severe combined immunodeciency (Scid)
in mice and defects lead to ionizing radiation hypersensitivity in humans.
ATM and ATR are found on meiotic chromosomes and have a role in
processing double-strand breaks generated during that process. These
molecules serve as sensors of ionizing radiation-generated DSBs. They not
only function as damage sensors but also as signaling molecules that relay
damage signals to regulatory proteins that halt the cell cycle progression
while repairs are made.
The ATM protein functions as a sensor of DNA double-strand breaks in
the following manner. In the absence of DSBs, the ATM proteins exist as
inactive dimers. Double strand breaks produced by, for example, ionizing
radiation, induce alterations in chromatin structure. The alterations in chromatin structure trigger the dissociation of the ATM dimers resulting in
autophosphorylation and activation. Once activated, ATM migrates over to
p53, to NBS1 and BRCA1, and to cell cycle checkpoint proteins, and phosphorylates them at one or more sites.
Figure 14.6. The cell cycle: Chevrons mark the divisions of the cell cycle into its four stages. The slash
placed late in G1 phase denotes the restriction (R)
point and the second slash placed in mitosis represents the spindle checkpoints. The three nonmitosis
stages are commonly referred to as interphase.
347
to produce two offspring. The three stages preceding mitosis are collectively
referred to as interphase.
Checkpoints are signaling pathways that ensure that a process does not
begin before a prior process is completed.They control the order and timing
of transition events in a network. DNA damage checkpoints, for example,
sense the presence of damage, producing signals that halt progression
through the cell cycle while the damage is repaired, and stimulate the transcription of repair genes to deal with it. The cell cycle includes several
checkpoints. The decision to undergo cell division or not occurs late in G1
phase. This point is called the G1/S checkpoint, or alternatively, START in
yeast and the restriction (R) point in mammals. Growth conditions signied by receipt of growth (mitogenic) signals from outside the cell are
evaluated, DNA is checked for damage, and a decision whether to proceed
through the cell division cycle is made. A second halt for checkpointing
occurs during S phase and the third takes place late in G2 phase and is
known as the G2 checkpoint. Checkpointing takes place during mitosis, as
well. The checkpoints halt mitosis if the spindle is damaged or if the chromosomes are not properly attached to the microtubules.
348
14. Cancer
Protein kinase
Cell-cycle phase
Cdk4, Cdk6
Cdk2
Cdk2
Cdk1 (Cdc2)
Cdk1 (Cdc2)
Late G1 phase
G1/S phase transition
S-phase
S-phase, G2-phase
G2-phase, M-phase
14.15 p53 Halts Cell Cycle While DNA Repairs Are Made
349
Figure 14.8. Growth signal stimulation of the transition from G1 phase to S phase:
Activated Cdk4s and Cdk6s along with kinases activated by growth signals hyperphosphorylate the retinoblastoma protein (pRb). In response, the E2Fs dissociate
from pRb and stimulate transcription of genes required for S-phase. Activation of
cyclin E is associated with the passage to S phase from G1 phase. The transcription
of cyclin E along with cyclin A and DNA polymerase is stimulated by the E2Fs.
These actions are terminated when Cyclin A-Cdk2 build up and turn off the E2Fs
transcriptional activities.
350
14. Cancer
Figure 14.9. Binding of an E2F peptide fragment to the pRb pocket: Shown in the
gure is a 18-residue fragment derived from the transactivation domain of E2F
(residues 410427) in a complex with the A/B pocket domain of pRb. The gure
was generated using Protein Explorer with atomic coordinates deposited in the
Brookhaven PDB under accession number 1N4M.
351
Figure 14.10. Cell cycle arrest triggered by DNA damage checkpoint signals: ATR
and ATM phosphorylate Chk1 and Chk2, which then phosphorylate the protein
phosphatase Cdc25 thereby inactivating it. Cdk2 is phosphorylated by the protein
kinase Wee1 and its actions are arrested. ATM signals through p53 to activate p21,
which arrests entry into S phase by inhibiting the transcriptional activities of the
E2Fs.
Figure 14.11. Controller circuit coordinating p53 and pRB activities: Mitogenic
signals are relayed into the cell via Ras, Ink4a and c-Myc. Pointed arrows denote
stimulatory inuences and at-headed arrows denote repressive inuences.
352
14. Cancer
disrupts formation of pRb-E2F heterodimers and thus promotes establishment of cancer cell cycles resembling those produced by mutations in pRb
itself.
The pRb and p53 work together. Early in the cell cycle, pRb inhibits the
E2Fs, and Mdm2 keeps p53 at a low level of activity. When pRb becomes
hyperphosphorylated its block on the E2F is relieved, ARF is stimulated
and it, in turn, stimulates p53 so that as the cell enters S phase, p53s surveillance activities are stepped up. If p53 detects DNA damage it signals
p21, which blocks the cyclin/cdk complexes from hyperphosphorylating
pRb. The retinoblastoma protein then inhibits the E2Fs while the repairs
to DNA are carried out.
The ATM/ATR and DNA-PKcs proteins activate p53 by stimulating
proteolysis of the Mdm2 inhibitor. Under normal cellular conditions, the
Mdm2 protein represses the checkpointing and apoptosis promoting activities of p53. The Mdm2 protein acts as a p53-specic, E3 ubiquitin ligase.
It suppresses the transcriptional actions of p53 by tagging it for proteolytic
destruction and keeping its expression levels low. One of the genes turned
on by p53 is the Mdm2 gene. This activity leads to the formation of an
autoregulatory loop in which p53 regulates Mdm2 at the level of transcription, and Mdm2 regulates p53 at the level of its activity. The Mdm2p53 system is balanced through these mutual dependencies so that the
appropriate signals can activate p53. These signals dislodge Mdm2 from
p53, leading to the transcriptional activation of p53.
353
Figure 14.12. Organization of the p53 protein: Shown in the upper part of the gure
is the domain structure of the p53 monomer. It contains an N-terminal transactivation (TA) domain, a proline-rich (PR) region, a DNA-binding domain, and in its Cterminal region a nuclear export sequence (NES), tetramerization (Tetra) domain
and a negative regulatory (NR) domain. Expanded depictions of some of the key
regulatory regions are shown in the lower part of the gure. The specic amino acid
residues that are phosphorylated (P), acetylated (A) or sumoylated (S) are shown.
Abbreviations: serine (S), threonine (T), lysine (K).
Figure 14.13. p53-DNA binding: The DNA-binding domains of the p53 protein are
depicted as ribbons while the dsDNA molecule is shown in a space-lled model.
The gure was generated using Protein Explorer with atomic coordinates deposited
in the Brookhaven PDB under accession numbers 1TUP and 1TSR.
354
14. Cancer
binding of the p53 molecule to the minor groove of DNA. Another often
mutated residue, Arg273, makes contact with the backbone phosphate.
The other residues help stabilize the interface between the p53 and DNA
surfaces.
355
progress towards more lethal states. Mutations in the p53 and pRb that
disable these proteins can contribute to the immortalization by disrupting
the conveyance of prosenescence signals.
Developmental Pathways
Fearnhead NS, Britton MP, and Bodmer WF [2001]. The ABC of APC. Human Mol.
Genet., 10: 721733.
Giles RH, van Es JH, and Clevers H [2003]. Caught up in a Wnt storm: Wnt signaling in cancer. Biochim. Biophys. Acta, 1653: 124.
Massagu J, Blain SW, and Lo RS [2000]. TGFb signaling in growth control, cancer
and heritable disorders. Cell, 103: 295309.
Peifer M, and Polakis P [2000]. Wnt signaling in oncogenesis and embryogenesis
A look outside the nucleus. Science, 287: 16061609.
Taipale J, and Beachy PA [2001]. The Hedgehog and Wnt signalling pathways in
cancer. Nature, 411: 349354.
356
14. Cancer
De Laat WL, Jaspers NGL, and Hoeijmakers JHJ [1999]. Molecular mechanisms of
nucleotide excision repair. Genes Dev., 13: 768785.
Hoeijmakers JHJ [2001]. Genome maintenance mechanisms for preventing cancer.
Nature, 411: 366374.
Kanaar R, Hoeijmakers JHJ, and van Gent DC [1998]. Molecular mechanisms of
DNA double-strand break repair. Trends Cell Biol., 8: 483489.
Lindahl T, and Wood RD [1999]. Quality control by DNA repair. Science, 286:
18971904.
Yang W [2000]. Structure and function of mismatch repair proteins. Mutation Res.
DNA Repair, 460: 245256.
Double-Strand-Break Repair
DAmours D, and Jackson SP [2002].The Mre11 complex:At the crossroads of DNA
repair and checkpoint signaling. Nature Rev. Mol. Cell Biol., 3: 317327.
DAndrea AD, and Grompe M [2003]. The Fanconi anaemia/BRCA pathway.
Nature Rev. Cancer, 3: 2334.
Hopfner KP, et al. [2002]. The Rad50 zinc-hook is a structure joining Mre11 complexes in DNA recombination and repair. Nature, 418: 562566.
Leuther KK, et al. [1999]. Structure of DNA-dependent protein kinase: Implications
for its regulation by DNA. EMBO J., 18: 11141123.
Song BW, and Sung P [2000]. Functional interactions among yeast Rad51 recombinase, Rad52 mediator, and replication protein A in DNA strand exchange. J. Biol.
Chem., 275: 1589515904.
Yang HJ, et al. [2002]. BRCA2 function in DNA binding and recombination from
a BRCA2-DSS1-ssDNA structure. Science, 297: 18371848.
Problems
357
Telomere Maintenance
Blackburn EH [2001]. Switching and signaling at the telomere. Cell, 106: 661673.
Chan SWL, and Blackburn EH [2002]. New ways to make ends meet: Telomerase,
DNA damage proteins and heterochromatin. Oncogene, 21: 553563.
De Lange T [2002]. Protection of mammalian telomeres. Oncogene, 21: 532540.
Itahana K, Dimri G, and Campisi J [2001]. Regulation of cellular senescence by p53.
Eur. J. Biochem., 268: 27842791.
Maser RS, and DePinho RA [2002]. Connecting chromosomes, crisis and cancer.
Science, 297: 565569.
Problems
14.1 Make a list of the ways a cancer cell differs from a normal one. Identify the signaling proteins that promote the altered response and how
these proteins are altered to promote cancer for each entry in the list.
For example, mutations in the Ras proteins that leave the GTPase
stuck in the on position contribute to aberrant growth signaling that
cannot be turned off.
14.2 Cancer is a multistep process. Arrange the entries in the list generated
in Problem 14.1 in a temporal order with early acting changes at the
top and late acting changes at the bottom. Which kinds of alterations
promote metastasis? Which ones help generate genome instabilities
and chromosome abnormalities?
15
Apoptosis
Kerr, Wyllie, and Currie coined the term apoptosis in an article that
appeared in the British Journal of Cancer in 1972. The term was taken from
the Greek and has the meaning of falling off as in the dropping of petals
from owers, and leaves from trees, which occur in a programmed manner
every autumn. Apoptosis, or programmed cell death, is widespread during
animal development where it is used to sculpt and rene tissues and organs.
Apoptosis makes possible the regression of tadpole tails, permitting the
emergence of an adult tailless form. It is used to remove larval organs in
insects, and to sculpt digitsngers and toesfrom undelineated limb buds
in mammals.
Apoptosis is widely encountered during development of the nervous
system. Some neurons are only transiently formed, and once their task is
completed they are removed. In many parts of the nervous system, cells are
initially overproduced. This overproduction ensures that there is adequate
input to target neurons during the initial phase of the nervous system connection. Excess cells are removed by numerically matching pre- and postsynaptic structures. The initial set of neural connections is rened, and cells
that are inappropriately wired are removed. In some neural populations,
most of the cells are removed in order to produce a set of precise neural
connections.
Apoptosis is an essential ingredient in the operation of the immune
system. Cells of the immune system such as B cells die off as they age and
when they are no longer needed. Virally infected cells, and damaged cells
that cannot be repaired, are targeted for apoptosis to prevent their harming
undamaged cells in the tissue or organ where they reside. The same strategy is used by the immune system to rid the body of cancer cells.
Apoptosis is different from necrosis. The plasma membrane does not
rupture in apoptosis as it does in necrosis, but instead cellular components
are degraded and packaged, and then digested. During apoptosis cells
undergo an orderly sequence of morphological changes. These changes
include cell shrinkage, chromatin condensation, DNA fragmentation, and
membrane blebbing, in which membrane-wrapped pieces of cell boil off of
359
360
15. Apoptosis
the cell surface as apoptotic bodies containing fragments of DNA and other
macromolecules.
Malfunctions in the cellular machinery that controls apoptosis are
encountered in many disease situations. Excessive apoptotic cell death
occurs in Alzheimers disease, Parkinsons disease, Huntingtons disease,
and ALS (Lou Gehrigs disease). Too little cell death is a hallmark of
cancer. In B-cell leukemia, for example, key regulators of the decision
circuitry that determines whether a cell survives or dies are overexpressed.
Apoptosis is suppressed and because population control is lost leukemia
develops.
361
362
15. Apoptosis
to L4 that determine the catalytic cleft are clearly visible in the left and
right hand panels of the gure. The caspases are still not fully activated in
the more open caspase form, but a nal set of changes occurring during substrate binding renders the caspases fully active.
363
Table 15.1. Mammalian caspases and their roles in the cell: Group III initiator
caspases act upstream of the Group II effector caspases.The executioners have small
prodomains and require assistance of the initiators for their activation. Four element
consensus sequences that the caspases recognize and cleave are presented in column
4. The most conserved residue in the consensus sequence is the Asp (D) residue
proximal to the cleavage site while the residue in the fourth position mostly
determines the substrate specicity.
Group
I: ICE
II: Effectors
III: Initiators
Caspase
Consensus
sequence
Prodomain
1
4
5
3
6
7
2
8
9
10
(WL)EHD
(WL)EHD
(WL)EHD
DExD
(ILV)ExD
DExD
DExD
(ILV)ExD
(ILV)ExD
(ILV)ExD
Large, CARD
Large, CARD
Large
Small
Small
Small
Large, CARD
Large, DED
Large, CARD
Large, DED
364
15. Apoptosis
Table 15.2. Bcl-2 family of apoptosis regulators: Listed are the numbers of amino
acid residues in the proteins.
Bcl-2 subfamily:
Antiapoptotic
A1
Bcl-2
Bcl-xL
Bcl-w
Boo
Mcl-1
size (aa)
172
239
233
193
191
350
Bax subfamily:
Proapoptotic
size (aa)
Bak
Bax
Bcl-xS
Bok
211
192
170
213
Bad subfamily:
Proapoptotic
size (aa)
Bad
Bid
Bik
Bim
Blk
Bmf
Hrk
Noxa
Puma
197
195
160
196
150
186
91
103
193
365
Figure 15.3. Structure of the antiapoptotic protein Bcl-xL: Shown in part (a) of the
gure is a ribbon diagram of the portions of the protein whose structure could be
determined through X-ray crystallography (i.e., highly disordered regions are not
included in the model). Gray-scale shadings highlight the four BH regions. The correspondence between BH regions and the eight a-helices is presented in part (b)
of the gure. The gure was generated using Protein Explorer from Brookhaven
Protein Data Bank entry 1AF3.
the cytoplasm. Bid is a sensor of death signals sent into the cell through the
death receptors, and is localized in the vicinity of the death receptors. Bim
is sequestered at microtubule associated myosin V motors where it awaits
activation by cytokine and other stress signals. Bmf is also immobilized at
myosin V motors where it responds to loss of cell attachment (anoikis)
signals. Two other BH3-only proteins, Bik and Blk, function in the endoplasmic reticulum as sensors of cellular stress. The remaining BH3-only
proteins are regulated at the transcriptional level. Noxa and Puma are
transcribed in a p53-dependent manner and may be regarded as DNA
damage sensors, while Hrk and Bim are upregulated in response to growth
factor deprivation and cytokine withdrawal.
366
15. Apoptosis
367
Alternative name(s)
Apo-1, CD95
CD120a
Apo-3
Apo-2, DR4
DR5
368
15. Apoptosis
Figure 15.4. Signaling through the DISC located at the plasma membrane:
Depicted are the main positive-regulating signaling elements. Homotrimeric TNF
ligands bind homotrimeric TNF receptors. In response, several adapter molecules
are recruited to the cytoplasmic portion of the receptors. The FADD adapter mediates recruitment and activation of the Caspase 8 pathway. The TRAF2 and RIP proteins mediate signaling and activation of the IKKs, which disinhibit NF-kB from the
IKKs. The TRADDs also signal to the JNK MAP kinase cascade.
369
apoptosis. The IKKs are the key point of convergence of a variety of regulatory signals triggered by cellular stresses. The IKKs are activated when
recruited to and phosphorylated at the DISC. They, in turn, phosphorylate
the IkBs, resulting in the activation of NF-kB. In their prosurvival mode,
the NF-kB dimers translocate to the nucleus where they stimulate transcription of negative regulators of not only DISC signaling but also of mitochondrial proapoptotic signaling elements. As a consequence, the balance
between pro- and antiapoptotic factors is shifted in favor of the antiapoptotic ones and apoptosis is prevented.
Recall from Chapter 9 that MAP signaling pathways convey stress (JNK
and p38) and growth (ERK) signals from the plasma membrane to the
nucleus where they inuence transcription of a different sets of target genes.
As shown in Figures 9.4 and 15.4, the JNK pathway begins in the DISC,
where the TRADDs recruit and activate MEKK1, the rst of the kinases
in the MAP kinase cascade. The last kinase in the cascade is JNK. Once
activated this kinase translocates to the nucleus where it phosphorylates
members of the AP-1 family of transcription factors. A similar set of signaling steps occurs in the p38 pathway.
Transcription factors such as AP-1 family members and NF-kB reect
cellular conditions and prior signaling events in their transcriptional
activities. Depending on the specic mix of coactivators and corepressors
present, subunit composition, and the set of residues that have been phosphorylated (and acetylated), these transcription factors will either promote
or inhibit apoptosis. For instance, the c-Jun transcription factor, an AP-1
family member activated at the end of the MAP kinase cascade, usually
functions as a transcription activator, but can also function as transcription
repressors when associated with corepressors. In NF-kB signaling, exibility of response is provided by variations in subunit composition. Depending on Rel subunit composition, NF-kB will either promote apoptosis by
expressing TRAIL receptors or inhibit apoptosis by expressing antiapoptotic survival factors.
370
15. Apoptosis
of the intermembrane space and the matrix. It is composed of voltagedependent anion channels (VDACs) located in the outer mitochondrial
membrane and the adenosine nucleotide translocator (ANT) situated in the
inner membrane. The PTPC also includes the peripheral benzodiazepine
receptors, creatine kinases, hexokinase II, and cyclophilin D.
The mitochondrial PTPC is the main target of Bcl-2 signaling and is the
main site of their pro- and antiapoptotic actions. If a molecule can cross a
membrane by means of simple passive diffusion, driven only by a concentration gradient, then that membrane is permeable to that molecule. In
apoptosis, the Bcl-2 proteins regulate the permeability of the mitochondrial membrane to the apoptosis-promoting molecules. Bcl-2 proteins act as
sensors of stress signals and as regulators of mitochondrial membrane permeability. They regulate membrane permeability by binding to and altering
the pores formed by the ANT and VDAC, and by oligomerizing and
forming pores by themselves.
BH3-only proteins stimulate the release from mitochondria of Cytochrome c and other apoptosis-promoting factors. The BH3-only protein
tBid, for example, promotes the permeability-increasing activities of Bax
and Bak proteins, stimulates the remodeling of the mitochondrial cristae,
and triggers the release of Cytochrome c. Members of the anti-apoptotic
branch of the family inhibit apoptosis by sequestering BH3-only proteins
thereby preventing their stimulation of Bax and Bad. There are two kinds
of BH3 protein actions. Bid-like BH3 proteins facilitate the oligomeriza-
371
tion of Bax and Bak, while Bad-like BH3 proteins bind Bcl-2s and displace
Bid-like peptides from them.
372
15. Apoptosis
Action
Required for assembly of the
apoptosome
Required for assembly of the
apoptosome
Proapoptotic regulator
Proapoptotic regulator
Early acting nuclear factor
Later acting nuclear factor
373
Figure 15.6. Apaf-1 and the apoptosome: (a) Apaf-1 molecule bound to
Cytochrome c and Procaspase 9Shown are a pair of WD-40 repeats in the Cterminal Y domain that bind Procaspase 9, and a pair of CARD domains in the Nterminal that bind Cyto c. A exible arm containing a CED4 homology motif that
binds dATP/ADP connects the N- and C-terminal regions to one another. (b) Fully
assembled apoptosome illustrating how the individual Apaf-1 molecules associate
into a sevenfold symmetric platform.
374
15. Apoptosis
375
376
15. Apoptosis
Figure 15.8. Mitochondrial PTPCApoptosome circuit: Perturbations of the mitochondrial permeability transition pore complex (PTPC) stimulates the release of
apoptosis-promoting factors such as Cytochrome c and counter inhibitor of apoptosis proteins (IAPs). These act at the apoptosome to activate Caspases 3 and 9.
377
378
15. Apoptosis
highly local and directional signaling similar to that occurring at chemical synapses takes place. One of the early events taking place in stressgenerated apoptosis is an increased permeability of the PTPC to calcium
entry. This event leads to increases in permeability to cytochrome c, resulting in its increased efux from the mitochondria.
Combinations of proapoptotic conditions such as oxidative stresses and
calcium signaling between the ER and mitochondria can initiate apoptotic
responses under condition where one or the other condition by itself
cannot. The cooperation between the two is mediated by a positive feedback loop in which ROS generated in the mitochondria promote increased
Ca2+ release from the ER. The calcium accumulates in the mitochondria
and triggers further increases in ROS production. This process generates
the progressive depolarization of the inner mitochondrial membrane and
increases membrane permeation leading to cell death.
The PTPC is not the only control point where Bcl-2 family proteins exert
their regulatory inuences. They also act at the endoplasmic reticulum
where they inuence the release of Cytochrome c occurs through their
modulation of calcium signaling. They help maintain homeostatic control
over movement of calcium into and between the two organelles (the ER
and the mitochondria), and mobilize calcium release. Proapoptotic Bax and
Bak proteins localize to both the endoplasmic reticulum and mitochondria
where they control calcium trafcking between the two organelles. In
the ER, Bax and Bak help initiate the apoptosis process by contributing
to Caspase 12 activation, and in the mitochondria they help activate
Caspase 7.
379
Figure 15.9. Regulation of apoptosis by p53: DNA damage signals are conveyed to
p53 in the form of posttranslational modications of specic residues such as phosphorylation on Ser46. In response, p53 (along with p63 and p73, not shown) stimulates transcription of apoptosis-promoting proteins acting at the DISC, PTPC, and
apoptosome. It also diffuses to the mitochondria where it interacts with antiapoptotic Bcl-2 family members to increase membrane permeability.
depicted in Figure 15.9, p53 can increase the numbers of propapoptotic proteins. These include death ligands and receptors at the DISC, the Apaf-1
scaffold proteins at the PTPC, and proapoptotic multidomain Bax, and
BH3-only proteins, at the apoptosome. Second, p53 can translocate to the
mitochondria where it can interact with the antiapoptotic Bcl-xL and Bcl-2
proteins to increase the efux of Cytochrome c from the mitochondria.
Recall from the last chapter that some of the most lethal mutations to p53
occur in its DNA binding domain. This domain contains the binding site for
attachment to the Bcl-2 proteins so these mutations not only destroy p53s
ability to bind DNA in the nucleus but also negate its ability to interact
with the Bcl-xL proteins at the mitochondria.
380
15. Apoptosis
Caspases
Boatright KM, et al. [2003]. A unied model for apical caspase activation. Mol. Cell,
11: 529541.
Chai JJ, et al. [2001]. Crystal structure of a procaspase-7 zymogen: Mechanisms of
activation and substrate binding. Cell, 107: 399407.
Chang DW, et al. [2003]. Interdimer processing mechanism of procaspase-8 activation. EMBO J., 22: 41324142.
381
Bcl-2 Proteins
Cheng EHYA, et al. [2001]. Bcl-2, BclxL sequester BH3 domain only molecules
preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol. Cell, 8:
705711.
Cory S, and Adams JM [2002]. The Bcl-2 family: Regulators of the cellular life-ordeath switch. Nature Rev. Cancer, 2: 647656.
Gross A, McDonnell JM, and Korsmeyer SJ [1999]. Bcl-2 family members and the
mitochondria in apoptosis. Genes Dev., 13: 18991911.
Letai A, et al. [2002]. Distinct BH3 only domain either sensitize or activate mitochondrial apoptosis, serving as prototypes cancer therapeutics. Cancer Cell, 2:
183192.
Moreau C, et al. [2003]. Minimal BH3 peptides promote cell death by antagonizing
anti-apoptotic proteins. J. Biol. Chem., 278: 1942619435.
Scorrano L, et al. [2002]. A distinct pathway remodels mitochondrial cristae and
mobilizes cytochrome c during apoptosis. Dev. Cell, 2: 5567.
DISC Signaling
Enari M, et al. [1998]. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature, 391: 4350.
Nagata S [1997]. Apoptosis by death factor. Cell, 88: 355365.
Scafdi C, et al. [1998]. Two CD95(APO-1/Fas) signaling pathways. EMBO J., 17:
16751687.
Weber CH, and Vincenz C [2001]. The death domain superfamily: A tale of two
interfaces? Trends Biochem. Sci., 26: 475481.
Apoptosome Assembly
Acehan D, et al. [2002]. Three-dimensional structure of the apoptosome: Implications for assembly, pro-caspase-9 binding, and activation. Mol. Cell, 9: 423432.
382
15. Apoptosis
Bratton SB, et al. [2001]. Recruitment, activation and retention of caspase-9 and -3
by Apaf-1 apoptosome and associated XIAP complexes. EMBO J., 20: 9981009.
Mitochondrial Physiology
Duchen MR [2000]. Mitochondria and calcium: From cell signaling to cell death.
J. Physiol., 529: 5768.
Jacobson J, and Duchen MR [2002]. Mitochondrial oxidative stress and cell death
in astrocytesRequirement for stored Ca2+ and sustained opening of the permeability transition pore. J. Cell Sci., 115: 11751188.
Kowaltowski AJ, Castilho RF, and Vercesi AE [2001]. Mitochondrial permeability
transition and oxidative stress. FEBS Lett., 495: 1215.
Marchetti P [1997]. Redox regulation of apoptosis: Impact of thiol oxidation status
on mitochondrial function. Eur. J. Immunol., 27: 289296.
Ricci JE, Gottlieb RA, and Green DR [2003]. Caspase-mediated loss of mitochondrial function and generation of reactive oxygen species during apoptosis. J. Cell
Biol., 160: 6575.
Problem
383
p53 Regulation
Flores ER [2002]. p63 and p73 are required for p53-dependent apoptosis in response
to DNA damage. Nature, 416: 560564.
Mihara M, et al. [2003]. p53 has a direct apoptogenic role at the mitochondria. Mol.
Cell, 11: 577590.
Vousden KH, and Lu X [2002]. Live or let die: The cells response to p53. Nature
Rev. Cancer, 2: 594604.
Cancer Therapy
Herr I, and Debatin KM [2001]. Cellular stress response and apoptosis in cancer
therapy. Blood, 98: 26032614.
Johnstone RW, Ruei AA, and Lowe SW [2002]. Apoptosis: A link between cancer
genetics and chemotherapy. Cell, 108: 153164.
Kaufmann SH, and Earnshaw WC [2000]. Induction of apoptosis by cancer
chemotherapy. Exp. Cell Res., 256: 4249.
Nicholson DW [2000]. From bench to clinic with apoptosis-based therapeutic agents.
Nature, 407: 810816.
Problem
15.1 Some cell types are more susceptible to apoptosis than others. What
classes of cells are especially responsive to apoptosis signals? What
kinds of cells send out proapoptosis messages as part of their normal
functions? Briey describe some of the ways a cell has of regulating
its own sensitivity to apoptosis.
16
Gene Regulation in Eukaryotes
The nding that metazoan genomes are as small as they are was remarked
upon in Chapter 1. In the introductory chapter, several aspects of eukaryotic cell organization that make this possible were noted. One of the most
important consequences of the changes in going from prokaryotic to
eukaryotic cell organization was creation of many different ways of regulating gene and protein expression, and for ne-tuning and specializing the
functions being performed. As a result of changes in the way the cells are
organized and gene expression and protein function are regulated, large
increases in complexity become possible without requiring large numbers
of additional genes. This central aspect will be examined in detail in this
chapter, which is devoted to the regulation of gene expression and protein
synthesis. Transcription will be discussed rst, followed by alternative splicing and translation.
Transcription in eukaryotes, even in the unicellular yeast, is more
complex than in prokaryotes because of the presence of chromatin. Recall
from Chapter 2 that eukaryotic DNA is highly condensed. Most of the chromatin is transcriptionally silent; that is, the DNA is tightly wound about the
histone core and promoter sites are not accessible. The molecular machinery responsible for transcription not only contacts and manipulates the
DNA molecule but also interacts with and alters the chromatin structure.
The molecular machines involved in modifying the chromatin structure and
transcribing genes are large. They can contain 50 or more subunits and have
molecular masses of several megadaltons. An example of how large these
machines may become is supplied by an examination of yeast cells. A set
of complexes that might be found in a typical yeast cell is presented in
Table 16.1.
The rst kind of complex is the basal (core) transcription machinery,
which includes RNA polymerase II and a set of general transcription
factors. It binds at the transcription start site and catalyzes the polymerization of RNA chains from DNA templates. This complex is commonly
referred to as the preinitiation complex (PIC). The second kind of protein
complex is exemplied in yeast by a set of about 20 proteins collectively
385
386
Molecular mass
(MDa)
Basal transcription
>40
Mediator
SWI/SNF complex
20
11
1
2
SAGA
15
Machine
Function
Transcription
driver (PIC)
Signal integrator
Chromatin
remodeler
Histone modier
called the Mediator. Complexes similar to the Mediator have been found in
all metazoans examined. They contain suppressor of RNA polymerase B
(another name for RNA polymerase II) proteins (Srb proteins) and Mediator proteins (Med proteins), and serve as intermediaries between the
transcription factors and the basal transcription machinery. The last
two complexes modify chromatin by chemomechanical means. The rst
(SWI/SNF) breaks and reforms histone-DNA bonds, while the second
(SAGA) alters the chemical afnity of histones for the DNA thereby either
promoting or inhibiting transcription through acetylation of the histone
tails. If all the contributions from the four complexes are combined the
result is a machine with about 90 subunits and a molecular mass of 7 MDa.
(A few subunits are shared by more than one complex but this changes the
additive result only slightly.) These machines and the underlying mechanisms are highly conserved from yeast to man.
387
Figure 16.1. Eukaryotic core promoter: Promoters are major endpoints of signaling pathways and have multiple sites for binding proteins. (a) The core promoter
contains several binding sites for components of the basal transcription machinery
to bind. The TATA box is located 25 to 30 bp upstream from the transcription start
site, Inr, centered about the +1 position. The TFIIB recognition element (BRE) is
located just above the TATA box while the downstream promoter element (DPE)
is located about 30 bp from Inr. (b) The rst event to occur is the binding of the
TATA box binding protein (TBP) to the TATA box, followed by recruitment of
TFIIB to the TBP and the BRE, followed by recruitment of RNA polymerase II
and other elements of the PIC.
boxes. Both the TATA box and the Inr can direct transcription initiation by
RNAP II. TATA box sequences are present in promoters for many abundantly transcribed genes. Housekeeping genes and genes encoding growth
factors and transcription factors often have TATA-less promoters. They
utilize the Inr for correct positioning of RNAP II at the start site. Some
promoters, lacking both the TATA box and the Inr, rely on the DPE for
positioning.
388
Figure 16.2. Eukaryotic promoter consisting of the core promoter plus proximal,
distal, and downstream regulatory sequences called responsive elements (REs): The
REs are represented in the gure as dark patches separated from the core promoter
and from one another by spacers. A pair of insulators, one at each end, bounds the
gene regulatory region.
provides sites for attachment of coactivators and corepressors and mediates long range enhancer-promoter interactions, is called a tethering element
(Figure 16.2).
Distal sites can be located either upstream of the core promoter or downstream, in between coding regions and even inside introns. These are the
sites where the long-range interactions between regulatory proteins and the
basal transcription machinery take place. The positively acting proteins that
bind the DNA are called enhancers and those that inhibit transcription are
called silencers. The distal regulatory regions are generally larger than the
proximal ones and supply binding sites for up to a dozen or so proteins.
Besides proximal and distal sites, a third kind of siteone that marks a
boundaryis called an insulator. Insulators are DNA sequences that mark
boundaries between independent sections of DNA. They protect the genes
they delineate from inuences outside the protected regions, serving as
blockers of enhancer functions or as barriers against elements that inactivate chromatin. When situated between an enhancer and a promoter the
insulators are able to block action of that enhancer on the promoter. They
also block the actions of gene silencers that act on the chromatin, thereby
marking the ends of chromatin domains (Figure 16.2).
Promoters are major endpoints of signaling pathways. Transcription
factors (TFs), the signaling elements that relay signals into the nucleus, bind
to responsive elements in promoters in a sequence-specic manner.The TFs
contain DNA-binding motifs or domains. These motifs enable the proteins
to bind to specic DNA sequences just as protein recognition modules
enable one protein to bind to another in an amino acid sequence-specic
manner. A typical promoter contains several responsive elements. This
architecture allows a number of transcription factors to bind and jointly
regulate transcription.
Each gene has its own promoter site consisting of the core promoter and
a number of responsive elements for transcription factors. A signaling
pathway that activates downstream transcription factors can regulate the
389
DNA-binding domains,
transcriptional activation domains,
dimerization domains,
protein-protein interaction domains, and
nuclear localization sequences.
In the above, the protein-protein interaction domains mediate their interactions with upstream elements of the signaling pathways. The nuclear
localization sequences include nuclear import sequences and/or nuclear
export sequences. The DNA-binding domains are modules designed to recognize specic nucleotide sequences. Recall that the edge of each base pair
is exposed on the outside of the DNA helix. The DNA-binding domains
recognize distinctive patterns of hydrogen bond donors and acceptors, as
well as hydrophobic patches along the edges located in the major and minor
grooves. As is the case of protein-protein binding interfaces, electrostatic
surface complementarity is accompanied by geometric complementarity,
that is, by geometric matching of the shape of the DNA helix and protein
surface. Protein-DNA binding interfaces were discussed in general terms in
Chapter 4. It was noted that some DNA-binding proteins bind the major
groove; others bind the minor groove; and some simultaneously contact
both. Besides hydrogen bonds and hydrophobic patches, salt bridges and
van der Waals contacts contribute to the establishment of good surface
complementarity.
There are a number of prominent protein-DNA binding motifs. One of
these is the helix-turn-helix (HTH) motif. Others include the helix-loophelix (HLH), zinc nger, and basic region-leucine zipper (bZIP) domains.
390
391
Figure 16.4. CREB bZIP dimer bound to DNA determined using X-ray crystallography: The basic region (residues 285 to 307 straddle and grip the DNA helix
while leucine zipper residues 308 through 336 form a dimerization interface. The
basic residues contact the major groove of the CRE region in the promoter. Protein
helices are depicted as ribbons and DNA molecules as balls-and-sticks. The gure
was prepared using Protein Explorer with atomic coordinates deposited in the
Protein Date Bank under accession number 1DH3.
are interdigitized with the leucines from the partner molecule to form a
short coiled-coil dimerization region, hence the name leucine zipper.
These proteins also contain a region rich in basic residues that interfaces
with the DNA. The basic region begins at a location seven residues Nterminal to the rst leucine in the leucine zipper domain. The overall
domain is referred to as a basic region leucine zipper (bZIP) domain; the
resulting bZIP dimer tightly grips the DNA molecule in the major groove
as illustrated in Figure 16.4 for the CREB transcription factor (CREB transcription factors will be discussed in Chapter 21). A number of transcription factors possess a basic region plus HLH and leucine zipper domains.
These DNA-binding proteins are referred to as bHLH-LZ proteins. The
bHLH-LZ and bZIP proteins form a variety of homeodimers and heterodimers with one another.
The nal category of DNA-binding proteins to be discussed is the zinc
ngers. These proteins utilize one or two zinc atoms to help form compact
domains of about 30 amino acid residues. The zinc nger motif is often
repeated, and each small module is organized about a central zinc atom.
Adjacent zinc nger modules form an overall DNA-binding domain whose
zinc ngers grip the DNA molecule. This motif is widely distributed among
eukaryotic transcription factors. The specifc composition of these modules
is variable but all contain zinc atoms and independently fold into small
compact structures as depicted in Figure 16.5.
392
Figure 16.5. Zinc ngers of the TFIIA protein in contact with DNA determined
using X-ray crystallography: The DNA-binding domain of the transcription factor
TFIIA has six zinc ngers each organized by a central zinc atom (indicated in the
gure by small lled circles). Protein helices are depicted as ribbons and DNA molecules as balls-and-sticks. The gure was prepared using Protein Explorer with
atomic coordinates deposited in the Protein Data Bank under accession number
1TF6.
393
Table 16.2. Nuclear receptors and their ligands: Common abbreviations for the receptors are given within the
parentheses.
Steroid receptors
Androgens (AR)
Estrogens (ER)
Cortisol (GR)
Aldosterone (MR)
Progestins (PR)
Nonsteroid receptors
Ecdysone (EcR)
All-trans retinoids (RAR)
9-cis retinoids (RXR)
Thyroid hormone (TR)
Vitamin D3 (VDR)
394
Table 16.3. Basal transcription machinery: Listed are the components, the number
of subunits in each component, their mass, and their role in initiating transcription
(discussed further in the main text).
Component
TFIID-TBP
TFIID-TAFIIs
TFIIB
TFIIF
RNA Poly II
TFIIE
TFIIH
Number of subunits
Function
1
12+
1
2
12
2
9
38
15250
35
30, 74
10220
34, 57
3589
sites. The minimal set of general transcription factors assembled at the promoter includes in order of recruitment TFIID, TFIIB, TFIIF, TFIIE, and
TFIIH. Another GTF, TFIIA, is sometimes recruited to the promoter, and
this (disposable) unit assists in TBP binding and stabilization. The assembly of the minimal set at the core promoter is depicted schematically in
Figure 16.1(b).
TFIID is believed to be the rst component of the transcription initiation complex to be recruited to the promoter site. It is the major sequencespecic GTF, and is a common target of TF activation domains. TFIID
contains the TATA box-binding protein (TBP), and a collection of TBPassociated factors (TAFs).The TBP subunit is a general transcription factor.
It recognizes the TATA sequence in its minor groove and is present in all
RNA polymerization operations. The TAFs are promoter-selective and
different mixes of these factors are present in different promoter sites.
Recruitement of TFIID triggers the recruitment of TFIIA and then TFIIB
followed by TFIIF bound to RNA polymerase II, and after that TFIIE, and
then TFIIH. These additional GTFs that follow establish protein-protein
contacts and link to the promoter site through the TBP. The TBP bends the
DNA and forms a saddle with its inner surface in contact with the DNA
molecule covering an 8-bp region of DNA.
395
Figure 16.6. Top view of RNAP II determined by means of X-ray diffraction measurements at 3.1 and 2.8 resolution: Ten of the subunits are shown in this gure.
Superimposed on this structure is a representation of a DNA molecule entering the
RNAP II, and an arrow indicating the direction of transcription. The gure was prepared using Protein Explorer with atomic coordinates deposited in the Protein Data
Bank under accession number 1I50.
As shown in Figure 16.6, the DNA enters the assembly through a cleft
formed by Rpb1 and Rpb2. The DNA is gripped by protein jaws but
cannot pass straight through the molecule because of a protein wall and
makes a right angle bend facilitating the addition of nucleotide triphosphates (NTPs). The NTPs enter through a funnel and pass through a pore
to reach the active site for polymerization. Exit of the nascent messenger
RNA chain is guided by several other structural elements, most notably by
rudder, lid, and clamp elements. Most of these functional elements belong
to the large Rpb1 and Rpb2 subunits. These subunits and the assignments
of functional roles to portions of each are depicted in Figure 16.7.
396
Figure 16.7. Functional roles of the large subunits of RNA polymerase II: (a) Rpb1
subunit and (b) Rpb2 subunit. The gure was prepared using Protein Explorer with
atomic coordinates deposited in the Protein Data Bank under accession number
1I50.
397
process for that site works in the following way. An HDAC acts on K9 to
remove an acetyl group. Next, a histone methyltransferase-specic for that
site catalyzes the covalent addition of a methyl group. Then, heterochromatin (adapte) protein 1 (HP1) binds to the methylated K9 site and blocks
transcription. The binding of HP1 to the methylated histone tail is shown
in Figure 16.8.
398
Table 16.4. Three families of ATP-dependent SWI chromatin remodeling complexes: AbbreviationsATP-dependent chromatin assembly and remodeling factor
(ACF); chromatin accessibility complex (CHRAC); chromatin organization modier
(Chromo); Imitation SWI (ISWI); nucleosome remodeling factor (NURF); remodels
the structure of chromatin (RSC); sucrose nonfermenting (SNF); switch (SWI);
Yeast (y); Drosophila (d); human (h).
Family
SWI/SNF
ISWI
Mi-2
Complexes
Recognition domain
Bromodomain
SANT domain
Chromodomain
399
Figure 16.9. Yeast Mediator complex: Shown are the three Mediator modules
The Srb4 head domain, the Med9/10 body domain, and the Gal11/Sin4 tail
domain, each named for prominent members of the module.
are 420 by 185 with distinct head, body, and tail regions formed by specic groups of subunits. The smaller coactivator required for Sp1 activation
(CRSP) complex, which consists of a subset of the ARC-L subunits, is
still 360 by 145 . Another mammalian Mediator complex, called
SMCC/TRAP (SRB- and MED-containing cofactor complex/thyroid
hormone receptor-associated protein), has about 25 members. These large
mediator complexes attach to the RNAP II CTD and form holoenzymes
with a total mass of about 1.5 MDa.
400
401
The second binding site is the branch site. This site is characterized by the
nucleotide sequence YNYURAY, where Y is a pyrimidine. The letter A
in Figure 16.11 denotes this site. The next portion of the RNA molecule is
the pyrimidine tract, which as its name suggests is a string of pyrimidine
nucleotides. The last regulatory unit is the 3 splice site characterized by
either a CAG sequence or a UAG sequence. Splicing is mediated by proteins that bind to particular stretches of DNA just as transcription is mediated by proteins that bind to promoter sites.
402
commitment stage (complex E). The next stage, the assembly of the U2
snRNP at the branch point, assisted by the auxiliary proteins, is referred to
as the presplicing stage (complex A). In the third stage, a preassembled
U4/U6 U5 complex is recruited to the U2 snRNP and these components
are joined by the U1 snRNP. All components of the spliceosome are now
assembled into an early splicing machine (complex B) and, as shown in
Figure 16.12, the 5 and 3 ends of the intron have been brought together
to form a loop.
Once the rst phase of the splicing process is complete, the catalytic
phaseearly splicing and late splicing (complex C)begins. In the catalytic
phase the spliceosome removes the intron and joins the anking exons. The
catalytic process involves several steps and is accompanied by extensive
conformational changes and rearrangements of the spliceosome. The U2,
U5, and U6 snRNPs form the catalytic core of the spliceosome. These components remain attached to the removed intron, while U1 and U4 leave the
spliceosome earlier. At the end of the catalytic stage the introns and Py
tracts have been removed leaving the selected set of exons joined together
in a mature mRNA molecule.
Transcription and splicing are coupled processes. Splicing begins while
the pre-mRNA is still being formed, during the transcription elongation
process, rather than after completion. As noted above, long introns separate rather short exons from one another. The coupling of transcription with
403
splicing helps bring together the exons in the correct manner over the large
distances and enables components of the transcription machinery to recruit
proteins required for splicing.
404
Figure 16.14. Crystal structure of the N-terminal region (UP1) of the hnRNP A1:
Each subdomain contains an RRM and the overall structure of the module is that
of an extended RNA binding surface. The crucial amino acid sequences are located
on the two middle beta strands of each subdomain. The gure was prepared using
Protein Explorer with atomic coordinates deposited in the Brookhaven Protein
Data Bank under accession number 1UP1.
405
Figure 16.15. Organization of the mature mRNA molecule: The coding region is
anked by a pair of noncoding regions, a 5 end cap, and a 3 end polyadenylate tail.
Molecular mass
(kDa)
eIF1A
eIF2
eIF2B
eIF3
eIF4F
17
126
277
330
291
eIF4B
eIF5
69
45
Function
Promotes Met-tRNA, 40S, mRNA binding
GTP binding protein, guides Met-tRNA onto 40S
GEF for eIF2
Binds to 40S, prevents 60S binding
Complex of eIF4E, eIF4G, and eIF4A; eIF4E
binds to the cap; eIF4G interacts with eIF3 to
augment binding of the preinitiation complex;
eIF4A is an RNA-dependent ATPase and an
RNA helicase
Facilitates ribosome-mRNA binding
Stimulates hydrolysis of GTP associated with eIF2
406
Figure 16.16. Assembly of the ribosomal 40S and 60S subunits at the mRNA molecule: At the end of the process the 40S subunit, 60S subunit, and Met-tRNA are
positioned at the AUG start codon. This positioning is guided by the eIFs with the
eIF4E subunit gripping the cap and the eIF4G subunit serving as a scaffold.
ary complex associates with the small (40S) ribosomal subunit to form a
43S preinitiation complex.
Next, the preinitiation complex attaches to the mRNA near its 5 end. An
initiation factor complex, eIF4F, binds to the m7GpppN cap and prepares the
way for the attachment of the preinitiation complex to the mRNA. Upon
attachment the preinitiation complex scans in the 5 to 3 direction for the start
AUG codon and halts upon arrival.Another initiation factor,eIF5,stimulates
the hydrolysis by eIF2 of GTP to GDP, resulting in the dissociation of the initiating factors from the assemblage leading to the attachment of the large
(60S) ribosomal subunit, and translation elongation can begin.
407
408
Figure 16.17. Structure of the human 4E-BP1 protein: The cap-binding protein
eIF4E, along with its 4E-BPs, is a critical control point for protein synthesis. The
seven identied phosphorylation sites are indicated along with the N-terminal
RAIP (Arg-Ala-Ile-Pro) motif, the C-terminal TOS (Phe-Glu-Met-Asp-Ile) motif
and the eIF4E binding site. The RAIP and TOS motifs are essential for proper phosphorylation by upstream kinases.
Structure of RNAP II
Cramer P, Bushnell DA, and Kornberg RD [2001]. Structural basis of transcription:
RNA polymerase II at 2.8 ngstrom resolution. Science, 292: 18631876.
Gnatt AL, et al. [2001]. Structural basis of transcription: An RNA Polymerase II
elongation complex at 3.3 resolution. Science, 292: 18761882.
Woychik NA, and Hampsey M [2002]. The RNA polymerase II machinery: Structure illuminates function. Cell, 108: 453463.
409
Chromatin-Modifying Enzymes
Boyer LA, et al. [2000]. Functional delineation of three groups of the ATP-dependent
family of chromatin remodeling enzymes. J. Biol. Chem., 275: 1886418870.
Eissenberg JC [2001]. Molecular biology of the chromo domain: An ancient chromatin module comes of age. Gene, 275: 1929.
Horn PJ, and Peterson CL [2002]. Chromatin higher order folding: Wrapping up
transcription. Science, 297: 18241827.
Narlikar GJ, Fan HY, and Kingston RE [2002]. Cooperation between complexes that
regulate chromatin structure and transcription. Cell, 108: 475487.
Vignali M [2000]. ATP-dependent chromatin-remodeling complexes. Mol. Cell Biol.,
20: 18991910.
Zeng L, and Zhou MM [2002]. Bromodomain: An acetyl-lysine binding domain.
FEBS Lett., 513: 124128.
Mediator Complexes
Asturias FJ, et al. [1999]. Conserved structures of Mediator and RNA polymerase
holoenzyme. Science, 283: 985987.
Boube M, et al. [2002]. Evidence for a mediator of RNA polymerase II transcriptional regulation conserved from yeast to man. Cell, 110: 143151.
Dotson MR, et al. [2000]. Structural organization of the yeast and mammalian mediator complexes. Proc. Natl. Acad. Sci. USA, 97: 1430714310.
Lewis BA, and Reinberg D [2003]. The Mediator coactivator complex: Functional
and physical roles in transcriptional regulation J. Cell Sci., 116: 36673675.
Malik S, and Roeder RG [2000]. Transcriptional regulation through Mediator-like
coactivators in yeast and metazoan cells. Trends Biochem. Sci., 25: 277283.
Taatjes DJ, et al. [2002]. Structure, function, and activator-induced conformations of
the CRSP coactivator. Science, 295: 10581062.
Theory
Levine M, and Tjian R [2003]. Transcriptional regulation and animal diversity.
Nature, 424: 147151.
Orphanides G, and Reinberg D [2002]. A unied theory of gene expression. Cell,
108: 439451.
Alternative Splicing
Cceres JF, and Kornblihtt AR [2002]. Alternative splicing: Multiple control mechanisms and involvement in human disease. Trends Genet., 18: 186193.
Faustino NA, and Cooper TA [2003]. Pre-mRNA splicing and human disease. Genes
Dev., 17: 419437.
Graveley BR [2000]. Sorting out the complexity of SR protein function. RNA, 6:
11971211.
Krecic AM, and Swanson MS [1999]. hnRNP complexes: Composition, structure,
and function. Curr. Opin. Cell Biology, 11: 363371.
Maniatis T, and Reed R [2002]. An extensive network of coupling among gene
expression machines. Nature, 416: 499506.
Smith CWJ, and Valcrcel J [2000]. Alternative pre-mRNA splicing: The logic of
combinatorial control. Trends Biochem Sci., 25: 381388.
410
Staley JP, and Guthrie C [1998]. Mechanical devices of the spliceosome: Motors,
clocks, springs, and things. Cell, 92: 315326.
Problems
16.1 Transcription factors and cofactors can promote or inhibit transcription in several ways. List some of these ways. For each way give the
location/site of action along the DNA, the specic interaction that
mediates the response, and the effect of the binding interaction on
transcription.
16.2 The translation initiation factor eIF2 is a GTPase. Several different
modes of action of GTPases were presented in Chapter 13. For
example, Ras acted one way as a switch while Cdc42 cycles continuously, and thus operates in a different manner from Ras. Which of the
several GTPase modes of operation does eIF2 follow?
16.3 One of the early events in apoptosis is the shutting down of the translation machinery. Give some examples of how the apoptosis regulatory machinery discussed in the last chapter might signal and regulate
protein synthesis through interactions of apoptosis signaling/regulatory proteins with the translation initiation factors.
17
Cell Regulation in Bacteria
Bacteria are not only highly efcient organisms, but they are also the unseen
majority wherever they exist. Bacteria are found in large numbers in the
open oceans (1.2 1029), in soils (2.6 1029), in oceanic subsurfaces (3.5
1030), and in terrestrial subsurfaces (~1 1030). Each human body hosts
some 1013 bacteria. One of the locales of high concentration is the gut. From
500 to 1000 different species of bacteria populate the human intestinal tract.
The greatest concentration of bacteria is in the colon where there are about
1011 bacteria per gram of bulk material. This number is far greater than the
amount of bacteria living on the surface of the skin. From 1000 to 10,000
bacteria live on each square centimeter of skin. These numbers yield a total
skin population of about 300,000,000 bacteria.
Most of the bacteria that reside in the gastrointestinal (GI) tract are
anaerobic, harmless, and even benecial. One of the species of resident bacteria is the Gram-negative bacterium Bacteroides thetaiontaomicron. This
bacterium acquires and hydrolyzes dietary polysaccharides that would
otherwise be indigestible. Bacteroides thetaiontaomicron possesses a large
number of genes that encode signaling proteins and gene regulators allowing the bacteria to communicate with and regulate their local environments.
A symbiotic relationship is established between bacterial ora and their
host environment. The bacteria stimulate vascularization and development
of intestinal villi and stimulate epithelial cells to secrete bacterial nutrients.
The bacteria generate simplied carbohydrates, amino acids, and vitamins.
Not all bacteria found in the GI tract are harmless. One of the occupants
is the Gram-positive bacterium Enterococcus faecalis. This is an opportunistic pathogen that occupies a variety of ecological niches including soil,
sewage, and water. Inside the GI tract, this bacterium causes uninary tract
infections and is highly resistant to vancomycin, the preferred antibiotic
for treating gram-positive bacterial infections. The propensity for this bacterium to harm its host is tied to genetic material acquired through horizontal gene transfer (HGT) from other bacteria. More than 25% of its
genome consists of DNA acquired from other species.
411
412
413
Function
36.5
150.6, 155.2
20 to 70
Figure 17.1. Organization of the bacterial promoter: (a) The primary DNA binding
regions are shown together with the transcription start site, InR. (b) Arrangement
of the subunits of RNA polymerase together with a catabolite activator protein
(CAP) transcriptional activator dimer.
polymerase. The alpha subunit contains two domains connected by a exible hinge. As shown in Figure 17.1b, each alpha subunit has an N-terminal
domain (aNTD) that interfaces the subunit with the beta subunits, and a
C-terminal domain (aCTD) that mediates interactions with the DNA promoter and with regulatory proteins. The core, written symbolically as a2bb,
has a total mass of about 380 kDa. There are several different kinds of sigma
factors, and their masses vary from 20 to 70 kDa.The sigma factors are DNA
sequence-specic binding proteins that recognize and bind the two specic
promoter sites listed in Table 17.2. The bacterial transcription-competent
RNAP holoenzyme is thus of the form a2bbs.
The association of a sigma factor and the core RNAP unit is transient.
Shortly after initiation of transcription, the sigma factor dissociates from
the core unit, which proceeds with transcript elongation. This dissociation
makes possible the attachment of a different sigma factor subsequent to
termination of the transcription event. If alternative sigma factors are
present in sufcient numbers, they will out-compete the initial sigma factor
for binding to the core unit. The core is thus reprogrammed to bind to an
alternative promoter site and transcribe a different set of genes.
414
Consensus sequence
Binding
5-TATAAT-3
5-TTGACA-3
AT-rich tracts
s
s, TFs
aCTD, TFs
415
Heat shock
s54 family
E. coli s , B. subtilis s
E. coli sS (s38)
E. coli sH (s32)
E. coli sE (s24)
B. subtilis sB (s37)
E. coli sF (s28),
B. subtilis sD
B. subtilis sH
B. subtilis sE
B. subtilis sF
B. subtilis sG
B. subtilis sK
E. coli sFecI (s19),
B. subtilis sV, sZ
E. coli sN (s54),
B. subtilis sL
Control function
Housekeeping, vegetative
growth
Stationary phase
Stress response genes
Stress response genes
Stress response genes
Genes for motility and
chemotaxis
Postexponential,
competence and early
sporulation genes
Early mother cell genes
Early forespore genes
Late forespore genes
Late mother cell genes
Genes for iron uptake,
antibiotic production,
virulence factors, outer
membrane proteins
Nitrogen xation, genes for
degradative enzymes
bacteria modify many of their internal activities in these cases using the
stationary phase sigma factor sS. This sigma factor stimulates physiological changes needed in the presence of nutrient deprivation and toxic
chemicals.
Many stressful conditions have to be dealt with by bacteria. Some conditions, like temperature extremes, osmotic stresses, and pH effects, are
related to the chemical and physical environment. These stresses are dealt
with by stress response sigma factors. High temperature is a representative
example of a stressful condition. When high temperatures are encountered
bacteria such as E. coli and B. subtilis use sH and sB to trigger the synthesis of heat shock proteins. These stress response proteins refold proteins
that have become partially unfolded due to thermal effects, and if the proteins cannot be refolded tag them for destruction. Sporulation and extracytoplasmic function (ECF) sigma factors are two more families of sigma
factors involved in controlling specic stress responses. The sporulation
sigma factors control the development of spores in B. subtilis in response
to starvation conditions. The ECF sigma factors control a diverse set of
responses (for example, iron uptake, heavy metal resistance, antibiotic production, and induction of virulence factors) mostly involving the remodeling of the transport machinery located in bacterial membranes. Finally,
some members of the s54 family are involved in control of nitrogen
metabolism while others are involved in regulating stress responses.
416
Table 17.4. Transcription factors, their actions, and typical cellular roles.
Family
AraC/XylS
ArsR
AsnC
Crp
DeoR
GntR
LacI/GalR
LuxR
LysR
MarR
MerR/SoxR
TetR/AcrR
Actions
Cellular role
Activator
Repressor
Dual
Dual
Repressor
Repressor
Repressor
Activator
Dual
Repressor
Dual
Repressor
417
418
Transcription activators bind to sites upstream of the -35 box. Some bind
in the vicinity of the UP element while others bind to sites further upstream.
Many of these transcription activators exert their inuences by binding to
aCTD. Because of its exibility, aCTD can bind to activators over distances
from -35 to -100 bp.Transcription factors that bind to distant upstream sites
are often referred to as transcription enhancers. The DNA molecule itself
has a certain intrinsic exibility. Sequences such as A-T tracts are especially
bendable. This exibility plus bending brought on by protein binding
enables activators to come into physical contact with subunits of the
holoenzyme. Transcription repressors bind to sites close to the transcription
start site. These proteins prevent the holoenzyme from initiating transcription either by sterically inhibiting access or by binding the holoenzyme so
tightly that it cannot release and start transcription.
Figure 17.2. Bacterial operon: Shown are ve genes, labeled A through E, controlled by a single gene regulatory region partitioned into repressor (O), core promoter (P), and enhancer (E) sections.
419
Function
LacI
Repressor
LacZ
LacY
LacA
b-galactosidase
b-galactoside permease
b-galactoside transacetylase
Description
Binds to transcription start site and part of the
promoter, inhibiting RNAP
Hydrolyzes lactose to glucose plus galactose
Transports lactose into the cell
Nonessential enzyme
420
Figure 17.3. Organization of the lac operon: (a) Constitutive expression of the lac
repressor protein (LacI) blocks transcription from the lac promoter when it binds
to the operator. (b) Binding of LacI is blocked by allolactose allowing cAMP-bound
CAP proteins to bind and stimulate transcription by the NRA polymerase (RNAP).
Glucose is the favored sugar and if it is plentiful the expression of proteins needed to metabolize alternative nutrients is inhibited. The LacI
repressor is constitutively expressed and under plentiful glucose conditions
binds downstream of the -10 and -35 sites and blocks RNAP from initiating transcription. When lactose is present in the environment, some of it
will enter the cell and, in the form allolactose, will bind to the LacI repressor. The lactose binding induces structural changes in LacI and the repressor is no longer able to block RNAP.
The lac promoter is a weak one. The sequences at the -10 and -35 sites
are not optimal for RNAP binding and the polymerase needs the binding
activities of an activator, the catabolite activator protein (CAP), to efciently
transcribe the structural genes in the operon. CAP forms a complex with
cAMP. When glucose levels are high the cAMP levels are low and the
complex does not form. When glucose is absent, the cAMP levels are high
enough for the CAP/cAMP complexes to form. The complexes bind at a
site upstream of the RNAP binding site (CAP by itself does not), where
they help RNAP to bind and start transcribing the genes. In sum, under the
combined actions of the LacI (negative regulation) and CAP (positive
regulation), the lac genes are transcribed when lactose is present and
glucose is absent (Figure 17.3).
The structure of LacI is well suited for controlled interactions with DNA.
As shown in Figure 17.4, two of its domains, the so-called Headpiece and
421
Figure 17.4. Crystal structure of the Lac repressor bound to a DNA operator determined using X-ray crystallography: Several domains of the Lac repressor are
involved in the interactions. The headpiece domain binds to the DNA in the major
groove while the Hinge helix binds DNA in the minor groove. The core domain is
shown broken down into NH2 and CO2 subdomains that mediate interactions with
inducers and repressors. One other domain, the tetramerization domain that mediates interactions with other LacI proteins, is not included in the gure, but can be
seen in other crystal structures. The gure was generated using Protein Explorer
with atomic coordinates deposited in the Protein Data Bank under accession code
1EFA.
Hinge helix domains, bind to the DNA in its major and minor grooves,
respectively. The core domain enables the repressor to be regulated by allolactose, which induces allosteric transitions that prevent the protein from
binding DNA.
Regulatory proteins such as CAP are able to bend DNA. This ability can
be seen in Figure 17.5 where a CAP dimer is shown bound to its regulatory
DNA regions. The bending of the DNA is quite pronounced, and by this
means transcription factors and coactivators can make transcription either
easier or harder.
422
Figure 17.5. Crystal structure of DNA bending by a CAP dimer determined using
X-ray crystallography: The gure was generated using Protein Explorer with atomic
coordinates deposited in the Protein Data Bank under accession code 1RUN.
Escherichia coli and Salmonella enterica, these genes belong to a large agellar motor regulon. The genes in this regulon are organized temporally
into three classesearly, middle, and late (Figure 17.6).
A variety of cellular and environmental conditions are factored into the
decision to begin biosysnthesis of the agellar motor. The corresponding
signals impinge on a master control point, namely the promoter for the
hCD master operon, from which the early genes hC and hD are transcribed. The transcriptional activation complex then regulates the transcription of middle genes. These genes encode structural proteins and
assembly regulators for the basal body and hook of the agella. The timing
of events is carefully controlled during the assembly process. Two regulatory proteins are expressed during the middle phase. One of these, FlgM,
binds to and inhibits the second, FliA (s28). The inhibition on s28 by FlgM
is relieved after the middle stage components of the agellar motor have
been assembled. The s28 regulator then initiates transcription of the late
genes including those for the chemotactic receptors Tar, Tsr and Aer, and
the suite of Che proteins discussed in Chapter 6.
423
Figure 17.6. Assembly of the agella motor: Environmental signals arriving at the
master controller stimulate transcription of early (Class I) genes. FlhC and FlhD
jointly stimulate transcription of middle genes from middle (Class II) promoters.
FliA (s28) and FlgM (anti-s28) regulate transcription from late (Class III) promoters. Transcription of late genes is inhibited until completion of the basal body
and hook assembly signied in the gure by the sending of a checkpoint signal.
bolic and reproductive strategy. It leaves the vegetative state and converts
itself into an inactive spore that can survive for long periods of time. As
indicated in Table 17.3 ve sigma factors govern the transition from vegetative growth to spore. The rst to come into play is sH. This sigma factor
is followed in sequential fashion by sF, sE, sG, and nally sK. As the genes
selected by these sigma factors are expressed the bacterium ceases exponential growth. Its plasma membrane invaginates, partitioning the cell into
large and small compartments, each containing a chromosome generated
during the last round of DNA replication. Once the septum is completed
the large compartment, the mother cell, engulfs the small cell, the forespore, producing a cell within a cell. A series of coating stages follow where
the forespore and its chromosome are protected, and lastly the spore is
released through lysis of the mother cell; that is, the rupture of the plasma
membrane/cell wall and the dissolution of the outer cell.
A number of signals, integrated together, govern the decision whether
to undergo sporulation (Figure 17.7). These signals converge on the transcription factor Spo0A. This protein acts at the end of a phosphorelay
system involving kinases such as KinA/KinB, a phosphorelay protein Spo0B
and an upstream transmitter Spo0F. Signals sent through the phosphorelay,
and associated protein phosphatases such as RapA, RapB, RapE, and
Spo0E include metabolic information, DNA status, and cell density. These
424
signals inuence the phosphorylation status of Spo0A. The Spo0A transcription factor, along with KinA and Spo0F, is upregulated by sH. The
Spo0A operon contains the genes for sF and sE, and starts the expression
of spore and mother-specic genes.
425
Figure 17.8. Asymmetric cell division in Caulobacter crescentus: A swarmer cell differentiates into a stalked cell, which then undergoes asymmetric cell division to
produce a swarmer cell and a stalked cell. CtrA gene expression suppresses DNA
replication. This master controller is shut down through regulated proteolysis at the
end of G1 phase and is started up again in the new swarmer cell near the end of S
phase.
replicated, the chromosomes move to opposite ends of the cell, and a new
agellum is formed at the other pole. In the next phase, G2, the cell divides
into a swarmer cell and a stalked cell.
The key element in the regulatory network that coordinates the morphological changes with the cell cycle progression is CtrA. This regulatory
protein is the counterpart to the Spo0A transcription factor discussed in
the last section. The CtrA master cell cycle controller controls a large
number of genes, in this case, about 25% of all the genes whose expression
varies in a cell cycle-dependent way in C. cresentus. Cellular localization,
proteolysis, and phosphorylation all have role in regulating the behavior of
CtrA. The controller is expressed in the swarmer, but not in the stalker. The
CtrA proteins that are present in the swarmer at the end of G1 phase
undergo proteolytic destruction and thus are absent in the S phase starker.
Pole proteins belonging to the agellum and chemotactic sensor are also
cleared by proteolysis. These and the subsequent regulatory processes are
facilitated by a number of two-component signal transduction systems that
localize to one or the other of the poles. One of these systems is DivJ/DivK.
This two-component system facilitates the localization of CtrA to the pole
that will produce a new agellum and upon cell division will belong to the
new swarmer. Another protein, the histidine kinase CckA phosphorylates
CtrA after DNA replication takes place, thereby activating CtrAs transcriptional activities. In response, CtrA regulates the transcription of genes
involved in agellar biogenesis, cell division, DNA methylation, and DNA
426
427
Collective behavior
Gram-negative bacteria
Photobacterium scheri
Vibrio harveyi
Proteus mirabilis
Serratia liquefaciens
Myxococcus xanthus
Chondromyces crocatus
Pseudomonas aeruginosa
Escherichia coli
Luminescence
Luminescence
Swarming, migratory rafts
Swarming, migratory rafts
Fruiting body
Fruiting body
Antibiotic resistance, virulence factors
Virulence factors
Gram-positive bacteria
Staphylococcus aureus
Streptomyces coelicolor
Bacillus subtilis
Streptococcus pneumoniae
Virulence factors
Antibiotics, differentiation
Antibiotics, genetic competence
Genetic competence
428
429
membranes. Instead, they are shipped out of the sender by ABC transporters, and diffuse over to and subsequently bind transmembrane receptors of the recipient. The latter receptors operate as the sensor components
of two-component signal transduction systems (Figure 17.10).
In some species (e.g., B. subtilis and S. pneumoniae) quorum sensing triggers a bacterial state called genetic competence that is geared for the uptake
of exogenous DNA. In others a threshold-exceeding density of cells induces
430
a virulence response (S. aureus) and the expression of virulence genes (P.
aeruginosa), or the formation of a biolm, a colony of bacteria collectively
exhibiting an increased resistance to antibacterial agents.
431
of these is conjugation, in which two cells establish direct contact with one
another. Sex pili are formed and genetic material in the form of a plasmid
is sent from the donor bacterium to the recipient. This is perhaps the most
common way of carrying out horizontal gene transfer. A second way of
transferring genetic material is through transduction, where a bacteriophage picks up genetic material from one bacterium and delivers it to
another. A third way of acquitting genetic material is through transformation, in which a bacterium becomes competent to capture exogenous DNA
from its local environment as discussed in the previous section.
Resistance to antibiotics can be spread rapidly through horizontal gene
transfer. One consequence of horizontal genetic transfer is that groups of
genes that endow one bacterial species with antibiotic resistance can be transferred in minutes to hours to another species.The transfer of genetic material,
that is, the sharing of blueprints for making useful proteins among bacteria, is
a particularly potent adaptive response.It is far more efcient to simply transfer a complete set of genes than to have each species invent for itself the
genes one by one by means of random mutations. In examining the complete
genome sequences of these pathogens it becomes clear that entire regions of
DNA have been acquired from other species in the past.
Antibiotics are agents synthesized by fungi and bacteria that kill competing microbes. There are several different kinds of antibiotics. Some
antibiotics act on bacteria by causing damage to their cell walls. Other
antibiotics impede the ability of the bacteria to carry out protein synthesis
or to disrupt the replication of DNA or to interefere with metabolism. Penicillin-type antibiotics such as Ampicillin and Methicillin, cephalosporin
antibiotics, and Vancomycin attack bacterial cells walls. Protein synthesis
impeding antibiotics include tetracycline, aureomycin, streptomycin, and
Erythromycin. Quinolones disrupt DNA replication, and sulfur drugs interfere with bacterial metabolism.
There are several ways of conferring resistance to antibiotics. One way is
to synthesize biomolecules that destroys the antibiotics. Another approach
is to pump the antibiotics rapidly out of the cell thereby keeping its intracellular concentration so low that it is ineffective. Still another way of
dealing with the antibiotic is to synthesize cellular agents that bind antibiotics and neutralize them thereby acting as intracellular blockers.
432
Its genome is 4.6 MBp long, while the pathogenic strain of E. coli O157 is
considerably larger5.4 MBp in length. In E. coli O157 there are O
islands and K islands, clusters of genes that encode a total of 1387 proteins not found in the K12 strain. The additional proteins encoded by the
E. coli O157 strain endow the bacterium with enterohaemorrhagic abilities.
One of the most striking ndings from sequencing complete genomes of
microbes is that bacterial genomes are riddled with phage genes. It appears
that bacteriophages are a major source of mobile genetic material moving
in and out of bacteria. In many species, pathogenicity and virulence of the
bacterial strain are tied to the presence of a prophage, the inactive (lysogenic) bacteriophage whose genes have been integrated into the bacterial
genome. An example of this coupling is provided by group A Streptococcus
(GAS) strains. These bacteria promote sore throats, impetigo, acute rheumatic fever (the major heart disease of children), necrotizing fasciitis (the
disease caused by esh-eating bacteria), and toxic shock syndrome. The
various strains (serotypes) of GAS are given M designations according
to differences in a family of streptococcus cell surface proteins called M
proteins, which impede the engulfment by phagocytes. The M18 strain,
responsible for acute rheumatic fever contains phage genes that encode the
disease-causing toxins. Another strain, M3, which produces toxic shock syndrome and necrotizing fasciitis, has a different set of phage genes from the
M18 strain, and the strain most often responsible for sore throats, M1, has
yet another set of phage genes.
Pathogenicity islands, or virulence cassettes, are clusters of functionally
related virulence genes that are absent in nonpathogenic organisms. The
term virulence denotes the ability to cause disease. A virulence factor is
the designation given to an individual disease-causing or disease-promoting agent of an organism. Some examples of virulence factors are toxins,
surface adhesion molecules, control molecules, and virulence regulatory
proteins (transcription factors). Virulence cassettes, consisting of ensembles
of these proteins, may be located on plasmids or on other mobile genetic
elements that can be transferred from one bacterium to another. They may
be found on the bacterial chromosome, as well. In all cases they encode a
complete suite of proteins needed to promote virulence.
The virulence cassettes of enteropathogenic E. coli (EPEC) strains allow
them to build their own signal transduction pathway in a host cell, which
they use to remodel the hosts cytoskeleton. The bacteria rst send soluble
proteins into the host cell membrane. The proteins are then modied to
form physiologically functional receptors for its bacterial ligands, thereby
establishing a rm contact between cells. Upon appropriating the communication machinery the bacterium uses the hosts resources to erect a
pedestal on the host cells surface. This pedestal supports the subsequent
assembly of a bacterial colony.
Virulence cassettes are used to transfer agents from bacterium to host.
The most common forms, known as Type III and IV secretion systems, are
433
Figure 17.11. Bacterial death modules: The genes for toxin and antitoxin (antidote)
reside side by side on a plasmid. When the genes are expressed the antidote binds
the toxin and prevent it from binding to its substrate. In response to suicide signals,
proteolytic enzymes degrade the antidote; the toxin then binds to its substrate
leading to cell death.
434
lived antidote that antagonizes the activity of the toxin. If the plasmid is
disabled the antidote will not be made, and because it has a short half-life
the antidote will decay away rapidly. When the antidote is removed either
by loss of the plasmid or by some other means such as proteolytic cleavage, the toxin is free to degrade its substrate, usually an enzyme critical for
cellular survival, and the host cell dies. It forming colonies, bacteria sometimes use death modules of this type to selectively remove a potion of their
population. This may be viewed as a bacterial form of programmed cell
death.
435
from the myxobacteria and act as quorum-sensing signals. If the concentration of A-factors is adequate, fruiting body formation proceeds.
A-signaling in M. xanthus is followed by C-signaling that determines
which cells in the nascent fruiting body are fated to become spores and
which cells will retain their original rod shape. Transmission of C-signals
occurs between cells that are in close, end-to-end contact. Transmission is
ineffective when neighboring cells lie side by side. The circuitry involved in
C-signaling closely resembles that used in Notch signaling in eukaryotes. Csignals transmitted between ends of cells activate FruA, a transcription
factor that stimulates the expression of the csgA gene that encodes the Csignals. This positive feedback loop increases FruA activity above thresholds for its activation of frz genes and then dev genes. The frz genes encode
proteins that mediate the aggregation of the cells into mounds, and the dev
genes encode proteins that promote the differentiation of rod cells into
spores. Because of the geometric constraint on C-signaling, cells in the
periphery never experience threshold-exceeding concentrations of FruA
while those in the nascent fruiting body reach critical signal densities.
436
bial mats found in the hot springs of Yellowstone National Park, and populate nearly all ecosystems.
Members of genera such as Calothrix and Anabaena can form differentiated structures. The structures take the form of long laments in which
nitrogen-xing heterocysts are interspersed at regular intervals among photosynthetic vegetative cells. The heterocysts reduce N2 to ammonia but do
not carry out photosynthesis or carbon xation. The vegetative cells, on the
other hand, perform photosynthesis and x carbon. The technical problem
being solved through multicellularity is the need to separate photosynthesis from the nitrogen-xing due to the inactivation by oxygen of the nitrogenase complex responsible for nitrogen-xing. The heterocysts possess a
protective envelope and turn off their photosystem II. They export inorganic nitrogen to their neighbors and import nutrients. About 1 in 10 to 20
cells in a lament is a heterocyst. Cell-to-cell signaling is used to establish
and maintain the pattern of cell differentiation just as it does in multicellular eukaryotes. Most strikingly, the heterocysts are terminally differentiated cells, like those in metazoan tissues.
437
of the root cells. A nodule is formed containing a plant cell-derived peribacteroid membrane. This membrane envelops the bacteria, which multiply
and differentiate into bacteroids. In some symbiotic associations the bacteroids differ greatly in morphology compared to their free-living forms,
becoming several times larger and Y-shaped in form. Within the nodules
the bacteroids and legumes exchange metabolites, which pass through pores
and channels in the peribacteroid and bacteroid membranes.
Initial plant bacteria-signaling events not only launch the nodule-forming
process but also confer specicity and lter out potentially harmful associations from helpful symbiotic ones. Flavinoids secreted by the plants bind to
NodD proteins situated in the cytoplasmic membrane of the bacteria. The
NodD proteins are members of the LysR family of transcription factors.They
serve as sensors of plant signals and as activators of genes whose promoters
contain a 49 Bp sequence called a nod box. Binding by favinoid-NodD complexes results in the activation of the NodA, NodB, and NodC genes common
to all rhizobia. They encode enzymes that catalyze the synthesis of the
lipooligosaccharide core, or backbone, of the nod factors. Structural nod
genes that are species-specic are expressed at the same time. These genes
encode enzymes that modify or decorate the nod factor backbones in a
manner that depends upon the structure of the avinoids.
Gene Organization
Busby S, and Ebright RH [1994]. Promoter structure, promoter recognition and
transcription activation in prokaryotes. Cell, 79: 743746.
Ptashne M, and Gann A [1997]. Transcription activation by recruitment. Nature, 386:
569577.
Sigma Factors
Cannon WV, Gallegos MT, and Buck M [2000]. Isomerization of a binary sigma-promoter DNA complex by transcription activators. Nature Struct. Biol., 7: 594601.
Gralla JD [2000]. Signaling through sigma. Nature Struct. Biol., 7: 530532.
Sharp MM [1999]. The interface of s with core RNA polymerase is extensive, conserved, and functionally specialized. Genes Dev., 13: 30153026.
Flagellar Assembly
Aldridge P, and Hughes KT [2002]. Regulation of agellar assembly. Curr. Opin.
Microbiol., 5: 160165.
438
Chilcott GS, and Hughes KT [2000]. Coupling of agellar gene expression to agellar assembly in Salmonella enterica, Servovar typhimurium. and Escherichia
coli. Micro. Mol. Biol. Rev., 64: 694708.
Kalir S, et al. [2001]. Ordering genes in a agella pathway by analysis of expression
kinetics from living bacteria. Science, 292: 20802083.
Sporulation
Burbulys D, Trach KA, and Hoch JA [1991]. Initiation of sporulation in B. subtilis
is controlled by a multicomponent phosphorelay. Cell, 64: 545552.
Fabret C, Feher VA, and Hoch JA [1999]. Two-component signal transduction in
Bacillus subtilis: How one organism sees its world. J. Bacterial., 181: 19751983.
Jiang M, et al. [2000]. Multiple histidine kinases regulate entry into stationary phase
and sporulation in Bacillus subtilis. Mol. Microbiol., 38: 535542.
Perego, M, Glaser P, and Hoch JA [1996]. Aspartyl-phosphate phosphatases deactivate the response regulator components of the sporulation signal transduction
system in Bacillus subtilis. Mol. Microbiol., 19: 11511157.
Antigenic Variation
Barbour AG, and Restrepo BL [2000]. Antigenic variation in vector-borne
pathogens. Emer. Infect. Dis., 6: 449457.
Quorum Sensing
Davies DG, et al. [1998]. The involvement of cell-to-cell signals in the development
of a bacterial biolm. Science, 280: 295298.
Fuqua WC, Winans SC, and Greenberg EP [1994]. Quorum sensing in bacteria:
The LuxR-LuxI family of cell density-responsive transcriptional regulators.
J. Bacteriol., 176: 269275.
Kleerbezem M, et al. [1997]. Quorum sensing by peptide pheromones and two-component signal transduction systems in Gram-positive bacteria. Mol. Miocrbiol., 24:
895904.
Schauder S, and Bassler BL [2001]. The language of bacteria. Genes Dev., 15:
14681480.
Van Delden C, and Iglewski BH [1998]. Cell-to-cell signaling and Pseudomonas
aeruginosa infections. Emer. Infect. Dis., 4: 551560.
Problems
439
Problems
17.1 Nonlamentous cyanobacteria solve the problem of separating photosynthesis from nitrogen-xing in a way different from heterocystous
cyanobacteria. The nonlamentous forms carry out photosynthesis
during the day and x nitrogen at night. The separation of metabolic
440
tasks is made possible by the presence of an internal clock that synchronizes the organisms metabolic processes with Earths 24-hour
rotation (circadian) period.
Circadian clocks are found in plants where they regulate the
opening and closing of plant leaves with respect to the daily cycle of
light and dark. They are found in protists, fungi, and animals, as well.
All share a common set of properties such as the use of feedback loops
and the introduction of time delays. How might a circadian clock be
set up in a bacterium? How might the mechanism(s) used to x the
period and phase differ from those employed by eukaryotes?
Suggested Reading
Dunlap JC [1999]. Molecular bases for circadian clocks. Cell, 96: 271290.
Ishiura M, et al. [1998]. Expression of a gene cluster KaiABC as a circadian feedback process in cyanobacteria. Science, 281: 15191523.
Xu Y, Mori T, and Johnson CH [2003]. Cyanobacterial circadian clockwork: Roles
of KaiA, KaiB and the KaiBC promoter in regulating KaiC. EMBO J., 22:
21172126.
Young MW, and Kay SA [2001]. Time zones: A comparative genetics of circadian
clocks. Nature Rev. Genet., 2: 702715.
17.2 Some bacteria swim from place to place using a rotating agellum at
the back end to provide a driving force in the uid medium. Swimming is not the only method exploited by bacteria for motility.Another
means of locomotion is gliding motility. Social bacteria use this type
of motion, sometimes called twitching motility, quite often. Describe
the components of a locomotion system that might allow a bacterium
to glide along a surface.
Suggested Reading
Wall D, and Kaiser D [1999]. Type IV pili and cell motility. Mol. Microbiol., 32: 110.
Wolgemuth C, et al. [2002]. How myxobacteria glide. Curr. Biol., 12: 369377.
18
Regulation by Viruses
Viruses consist of nucleic acid cores encased in protein sheaths. The protein
sheaths surrounding and protecting the nucleic acid cores are referred to
as capsids.Viral capsids enclose either DNA or RNA.As might be expected,
the viruses belonging to the former class are known as DNA viruses, while
members of the latter group are called RNA viruses. Some RNA viruses
covert their RNA to DNA, and from there pass through both a transcription and a translation stage to make new viruses. These viruses are called
retroviruses. Other RNA viruses do not convert their RNA to DNA.
Instead, their RNA functions as messenger RNA molecules that are used
directly in a translation stage to synthesize proteins.
Many viruses use lipids and carbohydrates acquired from their hosts to
make an envelope. They encase their caspsids in lipids and carbohydrates
picked up from plasma membrane or from organelles such as the nucleus,
ER, or Golgi apparatus. These viruses are referred to as enveloped viruses.
Glycoproteins are an important component of the viral envelopes. These
proteins mediate contact with and entry into the host cell through their
ability to bind to receptors on the hosts plasma membrane.
Viral genomes vary greatly in size. Some, like those of the picornaviruses,
are small, less than 10 kBp, and encode on the order of 10 proteins. Others,
such as poxvirus genomes, are large. They range in size from 130 to 300 kBp
and encode ~200 proteins, approaching those of the mycoplasmas in size of
their genomes. In all cases, the viruses require assistance and resources from
the host cell to replicate and propagate. For that reason their genomes
encode not only genes of a structural character that encode proteins for the
capsid and envelope, but also genes involved in replication and genes of a
signaling and regulatory character. The viral signaling and regulatory genes
encode proteins that regulate viral replication, disrupt and disable host
immune/protective responses, and interact with cellular systems in order to
use them for transport and replication.
In the rst part of the chapter the viral cycle of entry, replication, and exit
will be examined. This discussion will be followed by a general sketch of
some of the ways viruses deal with the immune system. The remainder of
441
442
the chapter is devoted to detailed discussion of how signaling and regulatory proteins make possible the survival of specic viruses. The hepatitis
C virus responsible for chronic hepatitis, cancer-causing viruses, human
immunodeciency virus (HIV)the causative agent of acquired immunodeciency syndrome (AIDS), and bacteriophages will be examined.
443
complex, their choice depending on the virus type and size. Virus particles
of diameter less than 30 to 40 nm are small enough to pass through the NPC
intact, without dissolution of the capsid, but larger viruses cannot pass
through without some rearrangement and capsid disassembly. These larger
viruses must either wait for dissolution of the nuclear envelope during
mitosis, or they must at least partially disassemble to pass through the NPC
during interphase.
Proteins that are actively transported into the nucleus are referred to as
being karyophilic. These proteins rely on importins and Ran GTPases for
passage through the NPC in the manner discussed in Chapter 11. These proteins have nuclear localization signals that interact with importins a and b.
Many viruses enter and leave the nucleus in the same manner as cellular
proteins. In the case of small viruses that can pass through the NPC intact,
the capsids contain proteins bearing nuclear localization signals (NLSs).
Herpes simplex virus Type 1 (HSV-1) is an example of a virus that is too
large to pass into the nucleus without disassembly. HSV-1 is an enveloped
virus that fuses with the plasma membrane of the host cell. The intact capsid
is released into the cytoplasm and is transported to the nucleus where it
docks. In order to gain entry to the nucleoplasm of its nondividing host it
expels its DNA directly into the nucleoplasm in a spring-loaded-like
fashion, leaving its icosahedral 125-nm wide capsid outside the nucleus.
This is not the only mechanism utilized by large viruses to get their DNA
into the nucleus. In the case of adenoviruses such as Ad2, the capsids are
rst disassembled and then the DNA is transported through the NPC into
the nucleus. This is accomplished with the use of NLSs resident on proteins
that chaperone the DNA through the NPC. Viruses such as HIV-1 utilize
yet another mechanism. These viruses are able to bind directly to the
nuclear pore, and their DNA can enter the nucleus without requiring the
assistance of importins.
444
In order to exit the cell, virus particles interact with the cellular machinery responsible for multivesicular body (MVB) biogeneisis. MVBs are a
type of endosome having a multivesicular appearance; they play a role in
receptor internalization, protein sorting operations, and viral budding. In
their role as devices for internalization and protein sorting, the MVBs transport receptors from the plasma membrane to lysosomes for either degradation or recycling back to the plasma membrane. These transport and
sorting operations are referred to as the MVB sorting pathway.
The proteins that participate in the vascular protein-sorting in MVBs are
called class E Vpss. The reason for this name is that, in the absence of
vascular protein-sorting (Vps) proteins, abnormal endosomes are formed
called class E compartments. The class E Vps proteins form complexes
called endosomal-sorting complexes required for transport (ESCRTs). In
humans, there are four distinct ESCRT complexes. Members of these complexes interact with the exiting viruses through late (L) domains. Late
domains are found in virus gag proteins in some viruses and in matrix proteins in others (virus gag and matrix proteins will be discussed shortly).
These L domains mediate interactions with the ESCRTs. The motifs found
in L domains are protein-protein interaction domains. The three most
common binding motifs are PXXP, PPXY, and YXXL. The rst of these
binds to proteins with SH3 domains; the second binds proteins possessing
WW domains, and the third provides attachment for clathrin-associated
adapter proteins. The last named motif is often found in cytoplasmic tails
of proteins involved in sorting and shipping.
445
Table 18.1. Viruses and some of the diseases they cause: AbbreviationsReverse
transcribing (RT); single-strand negative sense (-ss); single-strand positive sense
(+ss); double strand (ds).
Virus family
Type of virus
Adenovirus
Herpesvirus
DNA
DNA
Papovavirus
Poxvirus
Calicivirus
Filovirus
Flavivirus
Orthomyxovirus
Paramyxovirus
DNA
DNA
+ssRNA
-ssRNA
+ssRNA
-ssRNA
-ssRNA
Picornovirus
Rhabdovirus
Rheovirus
Togavirus
Lentivirus
Oncornavirus
+ssRNA
-ssRNA
dsRNA
+ssRNA
RT RNA
RT RNA
Diseases
Respiratory infections
Chickenpox, cold sores, encephalitis, genital
sores, Kaposis sarcoma, mononucleosis,
roseola, shingles
Warts
Cowpox, smallpox
Norwalk
Ebola, Marburg
Yellow fever, hepatitis C
Inuenza A and B
Measles, mumps, respiratory, colds (children),
pneumonia (adults)
Polio, hepatitis A, common cold
Rabies
Intestinal infections, respiratory infections
Rubella
AIDS
Human T cell leukemia
446
Cytokine signaling
Interferon signaling
from the viruses of the invaded cells. A third set of targets is the proteins
that regulate apoptosis. Cells infected by viruses are killed and induced to
undergo apoptosis. The viruses manipulate this protective response; in some
cases they turn off apoptosis and in others they use the process to hitch a
ride out of the cell. Finally, receptors bound in the plasma membrane are
targeted by viruses in order to gain entry into the cell.
447
At the most fundamental level, what is happening is that both host and
pathogen (viral and bacterial) have coevolved. Both have developed control layers tuned to the other, promoting adaptation and evolution of each.
The overall result is a balance, an equilibrium situation of coexistence of
host and bacterial and viral pathogens.
448
Figure 18.1. Organization of the genome of the hepatitis C virus: Proteins shown
as dark-shaded boxes are structural (S) in character while those in light-shaded
boxes are nonstructural (NS). From left to right the proteins are: Cnucleocapsid
protein, sometimes called the core protein; E1 and E2glycoproteins belonging
to the viral envelope; p7linker protein of unknown function; NS2 through NS5B
NS enzymes. Arrows denote cleavage sites. Solid arrows indicate sites cleaved by
host cellular signalases; the dashed arrow indicates the site cleaved by NS2, and the
dotted arrows denote sites cleaved by NS3.
family of viruses, which includes the yellow fever virus and several forms
of encephalitis-inducing virus.
The hepatitis C virus (HCV) contains a single strand positive sense RNA
(+ssRNA) molecule approximately 9.6 kBp in length that functions as a
messenger RNA. The genome encodes a single open reading frame that is
preceded at the 5 end by a noncoding region and terminated at the 3 end
by another such region. The resulting polypeptide chain, or polyprotein,
is proteolytically processed during and after translation into 10 proteins
(Figure 18.1). The four amino-terminal-most are structural in character. The
rst protein, C, encodes the capsid protein, and the next two, E1 and E2,
are glycoproteins that are inserted in the viral envelope. It is customary to
refer to proteins forming the virus capsid and envelope as structural proteins and proteins involved in replication and signaling/regulation as nonstructural. The fourth structural protein, p7, is a linker that separates the
structural from nonstructural proteins.
The nonstructural genes encode several different kinds of enzymes. The
NS5B gene encodes an RNA-dependent RNA polymerase that is responsible for viral replication. The NS2 and NS3 genes encode proteases that,
along with cellular proteases, chop up the large polyprotein into the structural and nonstructural proteins at the sites indicated in Figure 18.1. The
NS4B and NS5A gene products are regulatory in nature. The regulatory
gene products along with the E2 glycoprotein target the interferon system
of antiviral defense, allowing the viruses to survive and proliferate even
when that system is activated.
Recall from Chapter 9 that the cells rst line of defense to a viral attack
is the interferon system. During viral replication dsRNA is produced and
the detection of these molecules alerts the cell that viruses are present. A
sensor of dsRNA called protein kinase R (PKR) activates the interferon
response. PKR shuts off translation and stimulates transcription of a host
449
450
Figure 18.2. Retroviral control region: (a) A minimal retrovirus, containing gag,
pol, and env genes, is shown bounded at both ends by long terminal repeats (LTRs)
and a pX region near the 3 end. (b) An expanded view of the 5 LTR showing its
U3, R, and U5 regionsA further expanded view of the U3 region depicts a canonical set of DNA binding sites (responsive elements) for transcription factors. Abbreviations: cyclic AMP responsive element (Cre); glucocorticoid responsive element
(Gre); initiation, or start, site for RNA transcription (InR), located at the boundary
between U3 and R.
while the R and U5 regions are essential for viral replication. As illustrated
schematically in Figure 18.2, the U3 region is endowed with binding sites
for many of the key families of eukaryotic transcription factors. These sites
are capable of binding to both viral and cellular regulatory proteins.
A unique feature of HTLV-1 is the presence near the 3 end of its genome
of a region called pX that codes for several regulatory proteins. One of
these proteins, called Tax, binds to CREB proteins at the various Cre sites
not only in the viral genome but also in the cellular genome. Tax interacts
with a variety of cellular regulators of transcription including not only
CREB and cofactors but also NF-kB. Through its interactions with the cellular regulators, Tax changes the regulatory decisions of the T cells control
circuitry, immortalizing and transforming the cells.
451
Type of tumor
Liver cancer
Burkitts lymphoma,
nasopharylgeal carcinoma
Kaposis carcinoma
Cervical cancer
Hepatocellular carcinoma
Adult T cell leukemias
pesvirus (KSHV). Viruses such KSHV have pirated a large number of regulatory genes from their hosts, and through expression of these genes alter
the cell cycle and growth responses of the host so that they serve the virus.
They use the pirated genes, as well as genes that are unique to the viruses
to degrade both the innate immune response and the adaptive immune
response, and can transform cells, producing a number of cancers in
humans.
Some of the most prominent regulatory genes in the KSHV genome are
shown in Figure 18.3. The rst group of genes depicted in the gure encodes
452
two antiapoptotic proteins. The viral Bcl-2 protein acts at the PTPC to
inhibit apoptosis, and the viral FLIP acts at the DISC to throttle back death
receptor signaling by preventing activation of caspase 8. The next set of
gene products regulates the cell cycle so that it becomes more conducive
to viral replication. The viral cyclin protein (vCyc) is similar to the cellular
D cyclins and primarily binds to Cdk6. The cell cycle genes are followed in
the gure by a considerable number of genes that jointly regulate immune
responses and cell growth. The LAMP protein is a 12-pass transmembrane
protein that inhibits signaling from the B cell receptor. The other proteins
further shift the life-death signaling balance towards growth and away from
apoptosis, and suppress Th1 immune responses.
453
plasma membrane. They facilitate binding and entry of the virus into the
host cell. All retroviruses contain gag, env, and pol genes. The pol gene
encodes enzymes required for replication of the virus. One of the enzymes
is a reverse transcriptase (RT). This enzyme catalyzes the polymerization
of a DNA molecule from the RNA molecule. This is a reverse, or retro,
information transfer operation, hence the name retrovirus. The pol gene
also encodes a protease (PR) that cleaves viral proteins following their
translation, and an integrase (IN) that helps integrate the viral DNA into
the host genome. There are six regulatory genes. These genesvif, vpr, vpu,
nef, tat, and revare unique to HIV and along with gp120 are responsible
for HIVs lethal behavior. These virulence-promoting genes are discussed
next in greater detail.
454
occluded by the V1/V2 loop. The other overlaps the chemokine binding site
and is occluded by the V2 and V3 loops (Figure 18.6). This is not the only
technique used. A second way HIV avoids neutralizing antibodies is by conformational masking.This is a thermodynamic effect, tied to conformational
changes, that allows gp120 to bind to CD4 while simultaneously avoiding
neutralization by the antibodies. Yet another way used by gp120 to avoid
discovery is glycosylation. A large number of changing sugar groups are
added, about 20 glycans for each gp120 molecule. These act as a constantly
shifting shield against neutralizing antibodies.
455
molecules. In the early stages of HIV infection, short 2-kB mRNAs are produced and exported from the nucleus. These short transcripts encode Tat,
Rev, and Nef proteins, and the genes for these are referred to as early acting
genes. Later in the infection cycle longer 4-kB variants and full length
mRNAs are made, and the genes transcribed at that time are termed late
acting genes.
The HIV virus not only supplies its own promoter, as do other retroviruses, but also supplies its own transcription activator, Tat. The Tat protein
is an RNA binding protein. It binds to a segment of nascent HIV RNA
chain called the transactivating response (TAR) region located at the 5 end.
A number of cellular proteins recognize the TAR sequence and are
recruited to the site. As depicted in Figure 18.7, these include Cyclin T1 and
its cyclin-dependent kinase Cdk9, which hyperphosphorylates the Cterminal domain of RNAP II. In the absence of hyperphosphorylation, the
transcripts generated by RNAP II tend to be short. Hyperphosphorylation prevents premature elongation termination and enables the enzyme
to catalyze full length transcripts.
Rev is a sequence-specic RNA binding protein that binds to a region
called the Rev responsive region (RRE) located in env gene transcripts.
The Rev protein contains a nuclear export sequence (NES) and mediates
the export of all transcripts except those of tat, rev, and nef from the nucleus
into the cytoplasm, where they are translated into the virus components. In
the early stages of an HIV infection, before Rev levels are sufcient to
support its export function, transcripts longer than 2 kB that get made
collect in the nucleus. In the later stages of HIV infection, Rev facilitates
the export of longer and full length transcripts from the nucleus.
The Nef protein helps the virus to control signaling by its host and creates
an environment conducive to replication of the virus. One of the key targets
456
of Nef is the MHC-I complex on the cell surface. Recall that MHC-I complexes are used to present antigens derived from, for example, viruses, on
the surface where they can be recognized by killer cells of the immune
system. Nef stimulates the internalization of these receptors and directs
them to the Golgi. Another target of the Nef protein is the CD4 receptor.
These are internalized and sent to digestive compartments. The MHC-I and
CD4 receptors are not the only immune system signaling elements targeted
by Nef. It also downmodulates the CD28 coreceptor. All of these actions
help disable signaling between cells of the immune system, enabling
infected cells to evade detection and destruction. In macrophages, which
along with CD4 T cells are the main targets of HIV, Nef induces the release
of chemokines that attract resting T cells and help to transmit the virus to
uninfected cells.
457
Figure 18.9. Organization of the bacteriophage lambda genome and its regulatory
genes: The organization of the six regulatory genes and two noncoding operator
regions are depicted in the lower portion of the gure.
458
Their gene products control the decision between the lytic and lysogenic
life.
Figure 18.10. Operation of the phage lambda decision circuit: The central portion
of the regulatory circuit in shown in an expanded view in the lower portion of the
diagram. The lysogenic state is selected when CI binds to the operator sites and
represses transcription from the left and right promoters. The lytic state is chosen
when the Cro protein binds to the operator sites.
459
sites nearest the left and right promoters, thereby blocking transcription of
the other genes, and the phage remains a prophage. The cro protein product
is an antirepressor that shuts off cI when it is bound to the operator sites.
As shown in Figure 18.10, it favors the innermost sites in the left and right
promoters. When it binds to these sites it shuts off cI transcription while
allowing transcription of the other genes from the left and right promoters.
This circuit exploits both positive and negative feedback.
One of the most interesting aspects of the phage decision circuit is the
occurrence of cooperativity in the two operator regions. Binding of a
protein to a site in one of the operator regions increases the probability
that a second protein will bind to a nearby site that would otherwise be
unoccupied due to weak binding. The initial binding events thus serve to
recruit additional proteins to the regulatory regions where they jointly
control transcription. This type of control is not unique to bacteria or to
phages; it was encountered earlier in the chapter on eukaryotic gene
regulation.
The decision circuit operates under environmental control. The decision
circuit is sensitive to good versus poor growth conditions and to exposure
to ultraviolet radiation. The outcome of the race between lysogeny and lysis
is determined by the concentration of CII in the cell. These proteins act as
enhancers of cI transcription. The concentration of CII in a healthy cell is
usually maintained at a low level by the activity of host proteases that are
sensitive to glucose levels. When nutrients are plentiful, CII levels are low
resulting in a stable lytic state of replication and release of new phages.
The CII concentration under poor bacterial growth conditions is elevated
leading to CI levels that are high enough to send the phage into a lysogenic
state.
The phage can transition from lysogeny to lysis in response to DNA
damage arising from UV or ionizing radiation. RecA proteins are synthesized by the host in response to ultraviolet damage to the cellular DNA.
These proteins stimulate the repair of damaged DNA by inactivating the
LexA repressor. The LexA repressor is similar to the lambda repressor CI.
When phage lambda is present, RecA targets CI as well. There is an
increased expression of Cro proteins leading to a lytic stage where the virus
escapes from the UV damaged cell.
460
Figure 18.11. Phage genes incorporated into the genomes of several strains of
enterohaemorrhagic E. coli: The toxin genes StxA and StxB are situated in between
the replication and late antitermination (Q) genes on the left and lysis genes on the
right.
References
General Reading
Schaechter M, Engelberg NC, Eisenstein BI, and Medoff G [1999]. Mechanisms of
Microbial Diseases (3rd edition). Baltimore: Lippincott, Williams and Wilkins.
Walker TS [1998]. Microbiology. Philadelphia: W.B. Saunders and Company.
References
461
Hepatitis C Virus
Bartenschlager R, and Lohmann V [2000]. Replication of hepatitis C virus. J. Gen.
Virol., 81: 16311648.
Goodbourn S, Didcock L, and Randall, RE [2000]. Interferons: Cell signalling,
immune modulation, antiviral responses, and viral countermeasures. J. Gen. Virol.,
81: 23412364.
Katze MG, He YP, and Gale M, Jr. [2002]. Viruses and interferons: A ght for
supremacy. Nature Rev. Immunol., 2: 675687.
Large MK, Kittlesen DJ, and Hahn YS [1999]. Suppression of host immune response
by the core protein of Hepatitis C virus: Possible implications for Hepatitis C virus
persistence. J. Immunol., 162: 931938.
HIV-1
Boshoff C, and Weiss R [2002]. Aids-related malignancies. Nature Rev. Cancer, 2:
373382.
Emerman M, and Malim MH [1998]. HIV-1 regulatory/accessory genes: Keys to
unraveling viral and host biology. Science, 280: 18801884.
Gu YP, and Sundquist WI [2003]. Good to CU, Nature, 424: 2122.
Jacotet E, et al. [2000]. Control of mitochondrial membrane permeabilization by
adenine nucleotide translocator interacting with HIV-1 viral protein R and Bcl2. J. Exp. Med., 193: 509519.
Karn J [1999]. Tackling Tat. J. Mol. Biol., 293: 235254.
Kwong, PD, et al. [2002]. HIV-1 evades antibody-mediated neutralization through
conformational masking of receptor binding sites. Nature, 420: 678682.
Lum JJ [2003]. Vpr R77Q is associated with long-term nonprogressive HIV infection and impaired induction of apoptosis. J. Clin. Invest., 111: 15471554.
462
Marin M, et al. [2003]. HIV-1 Vif protein binds the editing enzyme APOBEC3G and
induces its degradation. Nature Med., 9: 13981403.
Sheehy AM, Gaddis NC, and Malim MH [2003]. The antiretroviral enzyme
APOBEC3G is degraded by the proteosome in response to HIV-1 Vif. Nature
Med., 9: 14041407.
Stevenson M [2003]. HIV-1 pathogenesis. Nature Med., 9: 853860.
Swigut T, Shohdy N, and Skowronski J [2001]. Mechanism for downregulation of
CD28 by Nef. EMBO J., 20: 15931604.
Swingler S, et al. [2003]. Nef intersects the macrophage CD40L signalling pathway
to promote resting-cell infection. Nature, 424: 213219.
Swingler S, et al. [1999]. HIV-Nef mediates lymphocyte chemotaxis and activation
by infected macrophages. Nature Med., 5: 9971003.
Wei XP, et al. [2003]. Antibody neutralization and escapes by HIV-1, Nature, 422:
307312.
Wyatt R, and Sodroski J [1998]. The HIV-1 envelope glycoproteins: Fusogens,
antigens and immunogens. Science, 280: 18841888.
KSHV
Damania B, Choi JK, and Jung JU [2000]. Signaling activities of gammaherpesvirus
membrane proteins. J. Virol. 74: 15931601.
Moore PS, and Chang Y [2001]. Molecular virology of Kaposis sarcoma-associated
herpesvirus. Phil. Trans. R. Soc. Lond. B, 356: 499516.
Bacteriophage Lambda
Bell CE, et al. [2000]. Crystal structure of the l repressor C-terminal domain provides a model for cooperative operator binding. Cell, 101: 801811.
Campbell A [2003]. The future of bacteriophage biology. Nature Rev. Genet., 4:
471477.
McAdams HH, and Shapiro L [1995]. Circuit simulation of genetic networks.
Science, 269: 650656.
Ptashne M, and Gann A [1997]. Transcriptional activation by recruitment. Nature,
386: 569577.
Shiga Toxin
OLoughlin EV, and Robins-Brown RM [2001]. Effect of Shiga toxin and Shiga-like
toxins on eukaryotic cells. Microbes Infect., 3: 493507.
Wagner PL, and Waldor MK [2002]. Bacteriophage control of bacterial virulence.
Infect. Immun., 70: 39853993.
Problems
18.1 Engineered viruses are being devised for use in cancer therapy. The
goal of researchers is to nd natural viruses, or create genetically
modied ones, that can kill cancer cells while leaving normal cells
unharmed. This main idea behind creation of these oncolytic viruses
is to take advantage of the altered signaling pathways in the tumors
by devising viruses that selectively target those cells.
Problems
463
The rst viruses to be used in this way are the adenoviruses. These
viruses and other like them stimulate host cells to pass through the
cell cycle. If the host cell does not pass into S phase the viruses cannot
replicate their DNA. Adenoviruses encode 30 to 40 proteins that are
expressed sequentially. First immediate early genes, then early genes
and nally late genes are expressed. The rst protein to be made is
E1A. This control protein binds to the pRB protein of the host cell
and inactivates it (Figure 14.11). The E1B protein is made soon thereafter. It binds to p53 and inactivates it, too. These binding and inactivation events ensure entry of the host cells into S phase.
Recall from Chapter 14 that p53 and pRb central cell cycle controller circuitry and are mutated in most cancers. The inactivation of
these genes is an important step in transforming the host cell into one
that supports replication of the adenoviruses. How would you engineer an adenovirus to kill cancerous cells possessing mutated p53 proteins while leaving normal cells unharmed?
18.2 Some cells are permissive with respect to a virus while others are not.
In a permissive cell, the virus can gain entry by binding a receptor and
once inside the cell the viruses can replicate themselves. In nonpermissive cells, there arent any receptors that can be exploited for entry,
and if they do the signaling pathways do not support completion of
the viral replication cycle.
One of the signaling pathways often altered in cancer cells is the
Ras signaling pathway. One of the downstream targets in this growthpromoting pathway is the antiviral agent PKR. As mentioned earlier,
when activated PKR shuts down proteins synthesis thereby preventing viral replication. Phosphorylation by one or more kinases acting
downstream from Ras represses this activity. How would you engineer
an oncolytic virus to kill cancerous cells possessing mutated Ras
proteins while leaving normal cells unharmed? Note: dsRNA viruses
often encode an anti-PKR protein.
19
Ion Channels
The plasma membranes of nerve and muscle cells differ from those of other
cells in the body. Whereas the potential across the plasma membrane of any
cell will change when the ion permeability across the membrane is altered,
only in nerve and muscle cells will the converse occur. That is, in nerve and
muscle cells, the ion permeability will change as a result of a change in membrane potential. This property is a consequence of the presence in the
plasma membrane of large numbers of ion channels that open and close in
response to changes in voltage. When an ion channel opens, up to 10 million
ions can ow though its conducting pore per second, generating a few
picoamperes (10-12 amps) of current. The presence of a few thousand of
these channels endows the plasma membrane with the ability to actively
conduct action potentials. Ion channels are responsible for all electrical
signaling, and regulate a variety of processes including osmotic balance,
hormone release, heartbeat, motor movement, learning, and memory.
Ion channels render the plasma membrane permeable to the passage of
certain inorganic cations (Na+, K+, Ca2+) and anions (Cl-). The channels are
composed of several subunits arranged in a way that creates a hollow pore
that permits the one-way passive diffusion of ions from either outside the
cell in, or inside the cell out. Some channels permit the passage of sodium,
while others allow potassium ions or calcium ions or chloride ions to pass
through. Ion channels are not open all the time, but rather open and close
in response to extracellular ligand (neurotransmitter) binding, intracellular
ligand binding, or to changes in membrane voltage. They possess one or
more gates that mechanically open and close their pores to the passage
of ions in response to the regulatory signals.
The rst part of the chapter is devoted to an examination of how membrane potentials arise and how action potential can be generated from
voltage-dependent changes in the permeability of the plasma membrane. A
number of key biophysical concepts such as channel gating, and voltagedependent activation and deactivation will be introduced along with the
Nernst and HodgkinHuxley equations. The second part of the chapter is
dedicated to the mechanisms that make properties such as gating possible.
465
466
These mechanisms are revealed by electron microscopy and X-ray crystallography studies of the atomic structure of the ion channel proteins.
Figure 19.1. Ion concentrations inside and outside the cell and membrane permeability: (a) Inside and outside are separated by an impermeable membrane
represented by the double dashed lines. (b) The membrane separating the two
compartments contains ion channels and is permeable to passage of potassium and
chloride ions into the cell and sodium ions out of the cell. Ion concentrations are
shown in units of millimoles per liter (mM/liter or mM).
467
RT [X]out
ln
.
zF
[X]in
(19.1)
[X]out
61.54
log
.
z
[X]in
(19.2)
468
Table 19.1. Values for typical concentrations of four main kinds of ions inside
and outside mammalian skeletal muscle cells are presented along with the
values for the equilibrium potential calculated from the Nernst equation.
Ion species
[X]out(mM)
Na+
K+
ClCa2+
145
4
123
1.5
[X]in(mM)
12
155
4
10-4
Ex(mV)
+67
-98
-92
+129
(19.3)
As is customary, the calcium contribution, which is small, has been neglected. In Eq. 19.3, pK, pNa and pCl are permeability constants. The appearance of their ratios in Eq. 19.3 takes into account the relative permeabilities
of the membrane to the different ion species. Typical resting potentials are
in the range -60 to -80 mV. These values are far closer to the potassium
equilibrium potential than to the sodium equilibrium potential. The closeness of the membrane resting potential to the potassium reversal potential
is a reection of a far greater permeability for potassium than for either
sodium or chloride. As their permeability ratios in the GHK equation
become negligible, the expression for the resting potential approaches that
of the Nernst equation for potassium.The reversal potential may be thought
of as supplying a driving force for changes in resting potential.
469
opening of still more sodium channels. This continues for only a short while.
The positive feedback of sodium channel openings stimulating the further
opening of sodium channels is terminated when the channels inactivate.
At this point, the potassium channels, which also open in response to
depolarization, start to dominate. The buildup of positive charge inside the
cell through the entry of sodium cations is now countered by a decrease in
positive charge through the exit of potassium cations. This slower activity
moves the membrane potential towards the reversal potential for potassium
ions. The opening of potassium channels continues for some time resulting
in the repolarization of the membrane, and contributing to the shut down
of the sodium channels. At the start of the cycle the resting potential is
about -60 mV. The membrane depolarizes to the extent that its potential
reaches positive values due to the sodium inux. It repolarizes to hyperpolarized values of more than -70 mV driven by the potassium efux and then
relaxes back to its resting potential. The hyperpolarization stage is important. It is needed to relieve the inactivation of the sodium channels, and
prepare the membrane for the next round of activation and action potential generation.
As shown in Figure 19.2, the time course of the ow of sodium ions
through their channels differs from that of potassium ions through their
channels. Sodium and potassium channels are both activated by depolarization, but the sodium channels open for a short time and then shut down
Figure 19.2. Time courses of sodium and potassium currents through a plasma
membrane: Plotted are relative permeability values for the two ion species, that is,
the number of open channels per unit surface area as a fraction of the peak number
open channels/unit area for each ion species. Time is measured from initiation of an
action potential by a threshold exceeding depolarization.
470
even if the membrane potential has not changed from its depolarized value.
That is, the sodium channels inactivate after being activated for a short time.
Potassium channels activate more slowly than sodium channels, reaching
their peak conductivity values after the sodium channels shut down.
However, once they reach their peak conductivity values, the potassium
channels remain open for some time. That is, potassium channels are
noninactivating.
To summarize, sodium channels launch and generate the rising part of
the action potential; positive feedback sustains it, and potassium channels
generate the slowly declining following portion of the pulse. The positive
feedback loop of the sodium channels is the key to generation of action
potentials that propagate down axons without diminishing. Once a small
depolarization is started in a limited region of space, the spread of ions laterally to neighboring spaces triggers a growing depolarization in that space
and from there to the next neighboring space and so on. The pulse does not
die out since a small depolarization triggers a large one, that is, the signal
is amplied, as a result of the positive feedback. Thus, once started, a pulse
propagates along without losing strength, that is, the pulses are regenerative. The existence of a threshold all-or-nothing response is a consequence
of the positive feedback from the sodium channels and negative feedback
from the potassium channels. Negative feedback is present because the
depolarization-induced exit of potassium hyperpolarizes the membrane,
thereby acting negatively to shut down the potassium ion ow. As a result
of both forms of feedback there is an all-or-nothing response. It takes a
certain amount of depolarization to trigger the positive feedback dominated sodium inux, and once started the process runs to completion.
471
the squid giant axon. In these studies, a method, the voltage clamp, was
developed that allowed researchers to directly measure current ow across
the membrane of the axon.
The basic elements of the HodgkinHuxley model are a set of ionic currents separated into distinct contributions from sodium and potassium ions,
and from the leakage current, representing the fairly constant and voltageindependent, background leakage of ions through the plasma membrane.
These three currents are related to membrane permeabilities, which are
expressed in terms of ionic conductances gNa, gK and gL, and the displacements of the membrane potential from the reversal potentials for the ion
species. The relationship between current, conductance, membrane potential, and reversal potential is from Ohms law,
I x = gx (Vm - Ex ).
(19.4)
In the above, X denotes the ion species under consideration, Vm the membrane potential, Ix is its current, Ex is its reversal potential, and gx is its conductance, the inverse of its resistance Rx, that is,
gx = 1 Rx .
(19.5)
In elementary circuits, R is a constant, and Ohms law says that current and
voltage are linearly related. This is not the case for a biological membrane.
Although the linear form of Ohms law is retained in the HodgkinHuxley
equations, the sodium and potassium conductance are no longer constants,
but instead are treated as functions of the membrane voltage.
Because of the nonconducting nature of the lipid bilayer the plasma
membrane can be thought of as a capacitor that permits a buildup of charge
on its surfaces. If a current is applied part will contribute to charging up the
plasma membrane and part will ow through the membrane into the cell.
The net ionic current is the sum of the sodium, potassium, and leakage currents plus any external applied or synaptic currents. The capacitive current
IC is, from Faradays law,
IC = C
dVm
.
dt
(19.6)
In this expression, C is the capacitance, and dVm/dt is the time rate of change
in the voltage. The membrane voltage does not instantaneously adapt to the
ow of charge in and out of the cell. Instead, it takes a certain amount of
time for charge to build up on the membrane and the voltage to adjust to
the current ow. The complete circuit equation, expressing the conservation
of charge, is
C
dVm
= - g K (Vm - EK ) - g Na (Vm - ENa ) - g L (Vm - EL ) + I ,
dt
(19.7)
472
473
(19.8)
(19.9)
The quantities m h and n represent the fractions of open sodium and potassium channels, respectively, and the g-bars are constants representing the
474
475
Table 19.2. The four families of ion channels found in the plasma membrane of
neurons: Except for the outward directed potassium channels, ion channels allow
for the inward selective diffusion of the ion species listed in the second column.
AbbreviationsHyperpolarization-activated cyclic nucleotide gated (HCN);
chloride channel of the CLC family (ClC); nicotinic acetylcholine receptor
(nAChR); g-aminobutyric acid Type A, Type C (GABAA,C); 5-hydroxytryptamine
Type 3 (5-HT3); a-amino-3-hydroxyl-5-methyl-4-isoxazole propionate acid
(AMPA); N-methyl-d-aspartate (NMDA).
Ion channel
Selectivity
6TM cation
Calcium
HCN
Potassium
Sodium
Subunit topology
Channel assembly
6TM, loop
24TM, 4 loops
Tetrameric
Monomeric
18IM
Dimeric
4TM
Pentameric
3TM, loop
Tetrameric
Ca2+
Na+, K+
K+
Na+
Voltage-gated anion
ClC
Cl-
Cys-loop receptor
nAChR
GABAA,C
Glycine
5-HT3
Cations
Anions (Cl-)
Anions (Cl-)
Cations
Glutamate receptor
AMPA
kainate
NMDA
Na+, K+
Na+, K+
Ca2+
476
Figure 19.5. Ion channel subunit threading patterns: (a) Voltage-gated potassium
channelTransmembrane helices 5 and 6 along with the indicated loop form a
conducting pore. The positively charged residues in transmembrane segment 4 plus
portions of transmembrane helices 13 form the voltage sensor. (b) Bacterial ClC
showing the threading of 18 alpha helices of varying lengths. (c) Acetylcholine
receptor ion channel indicating the presence in the amino terminal portion of a
conserved Cys loop near the membrane surface. (d) AMPA receptor ion channel
showing the locations of S1 and S2 segments, which, along with the Flop segment,
form the ligand-binding domain.
477
Figure 19.6. Single chain voltage-gated cation channel topology: Shown in the
gure is a single 6 4-pass polypeptide chain topology characteristic of bacterial
KscA potassium channel.
rowest part of the pore) that allows certain ion species to ow through. In
channels that are voltage-gated, transmembrane segment 4 (TM4) contains
a number of positive changes (basic arginine or lysine amino acid residues),
and along with elements of TM13 functions as the voltage sensor. In channels whose gates are controlled by intracellular ligands, binding sites for the
regulators are location on the C-terminal segment that follows TM6.
Voltage-gated cation channels are assembled in one of two ways. In the
Shaker family of voltage-gated potassium channels, four of six TM core
units are arranged into the ring. In many voltage-gated sodium channels
and voltage-gated calcium channels, the amino acid residues replicated
among the four subunits form domains belonging to a single, long (2000
amino acid residues), 6 4 = 24-pass, polypeptide chain. In either assembly
mode, the result is a fourfold symmetric ring structure in the plasma membrane with a pore in the center. A typical threading pattern for a 24-pass
transmembrane protein, the bacterial KscA potassium channel, is presented
in Figure 19.6.
The threading pattern and assembly plan for voltage-gated anion channels are radically different from those of the cation channels. As shown in
Figure 19.5(b), the polypeptide chain of a ClC chloride channel subunit consists of 18 distinct alpha helices of varying lengths. Some of the helices pass
through the membrane, but others do not and one helix resides entirely in
the cytoplasm. As will be discussed shortly, the anion channel is formed by
two of these subunits, and in place of a single pore in the center there are
two off-center pores.
The two families of ligand-gated ion channels can be further divided into
several smaller families according to the types of ligands, or neurotransmitters, being bound. Some ligand-gated ion channels permit the passage
into the cell of cations, while others allow anions such as chloride to enter
the cell.When an anion enters the cell the net charge inside the cell becomes
more negative and the membrane voltage shifts to more hyperpolarized
values. Since action potentials are generated by threshold-exceeding depolarizations, the entry of negative ions such as chlorine suppresses ring.
Neurotransmitters such as GABAA,C and glycine that act through anionic
receptors are referred to as inhibitory neurotransmitters. Conversely,
478
479
Figure 19.7. Potassium channel voltage sensor: In the absence of depolarizing currents, the membrane is hyperpolarized. The positive charges in helix S4, along with
the other components of the S1 to S4 voltage sensor, maintain the channel in a
closed conguration. When depolarizing currents are present the S1 to S4 helices
move towards the outer leaet, and as a result the pore helices S5 and S6 shift their
orientation, thereby opening the pore.
480
481
Figure 19.9. The ClC chloride ion channel determined by X-ray crystallography: A
side view of the dimeric chloride-selective ion channel is presented. The approximate locations of the selectivity lter in the two subunits are denoted by the circled
Xs. The gure was generated using Protein Explorer with atomic coordinates
deposited in the Brookhaven Protein Data Bank under accession code 1KPK.
of the structural properties of the subunits. Their extracellular amino terminal contains a cys-loopa pair of disulde-bridged cysteines separated
by a stretch of 13 amino acid residues.
Nicotinic acetylcholine receptor ion channels are responsible for excitatory signaling at the connections between nerve and muscle cells, or neuromusclular junctions, located throughout the body. There are two types of
acetylcholine receptor ion channelsnicotinic and muscarinic. The former
bind to the nicotine whereas the latter binds to the muscarine, like nicotine
an alkaloid. Those nicotinic receptors, located at neuromuscular junctions,
are termed muscle-type nAChRs, while those found at neuron-to-neuron
contacts in the spinal cord ganglia and in the brain are referred to as neuronal nAChRs. As their name indicates, the neuronal nicotinic receptors
mediate nicotine addiction in smokers, whereas muscle-relaxing, paralyzing, and spasm-promoting toxins target neuromuscular nAChRs. Examples
of toxins that target nicotinic acetylcholine receptors are atropine, curare,
and scopolamine, all from plants, and cone snail and cobra toxins.
The nAChRs are pentamers, assembled in a symmetric or nearly symmetric ring with a pore in the center. There are 17 known vertebrate subunits. These subunits belong to one of ve groupsa, b, g, d, and e. Neuronal
receptors are typically homopentamers of a particular a subunit or heteropentamers of a and b subunits. The muscle-type of nAChR is less varied.
These receptors consist of two a1 subunits, plus one each of a b1, d, and g
or e subunit.
Perhaps more has been learned about the acetylcholine receptor than
any other neurotransmitter-gated ion channel. It has been studied with
482
483
tical AChBPs. The pore formed by the ve subunits is clearly visible in this
gure. As might be expected the residues lining the inner wall of the pore
are highly hydrophilic, and many are charged. A ligand-binding site is
present in a cavity formed at the interface between each adjacent subunit.
The cavity, positioned towards the outside of the ring, is formed by a set of
loops from one subunit and a series of beta strands from the other unit.
Since there are ve interfaces, the ligand binding domain can bind up to
ve ligands.
484
Glutamate receptor ion channels are responsible for most of the excitatory signaling in the brain. These receptors are most likely homo- and
heterotetrameric assemblages of subunits. They assemble in several stages.
Individual subunits rst associate to form dimers, and these dimers then
associate to form dimers of dimers. Like the voltage-gated ion channels, the
receptor ion channels have open and closed conformations. Ligand binding
results in stabilization of the open conguration through an allosteric mechanism. The receptors do not remain open indenitely, but rather desensitize and go back to a closed conformation soon (within milliseconds) after
ligand binding.
The S1S2 ligand-binding domain of an AMPA receptor is depicted in
Figure 19.11. The S1S2 domain depicted in this gure is a laboratory
construct made by cutting the polypeptide chain and then joining together
the S1 and S2 + Flop segments using a 13-residue-long linker. The result
is a ligand-binding domain that replicates the actions of the natural extracellular unit without the encumbrances of the other loops and transmembrane segments. As can be seen in Figure 19.11 the S1 and S2 semidomains
form a pair of jaws that grip the ligand. In the absence of ligand binding,
the jaws are open, but they close upon binding the ligand. The amount of
jaw closing is variable, and depends on which of several different kinds of
ligands are being bound. Binding by antagonists stabilizes the S1S2 domain
in an open conguration; the jaws partially close in response to binding
by partial agonists such as kainite, and close more fully in the presence of
glutamate.
The jaws are mechanically coupled of the gate. A linker that connects the
jaws to the membrane-spanning helices provides the mechanical coupling.
In the dimeric form, there are two linkers, one for each subunit. The linkers
move apart from one another as the jaws close and move nearer one
another when the jaws open. This movement opens and closes the gate.
When the jaws are open the gate is closed and when the jaws close through
agonist binding the gate opens.
485
Ions ow into the cell when the gate opens. The gate does not remain
open for very long, just a few milliseconds. The gate then closes even though
the ligand remains bound. The closing of the gate is brought on by
rearrangements of the dimer interface. The interface between subunits in
the dimer is placed under strain by the gate opening. The rearrangements
of the dimer interface relieve this strain by disengaging the conformation
changes brought on by agonist binding and shutting the gate, resulting in
receptor desensitization (to the ligand binding).
486
Problems
19.1 Using the Nernst equation with R = 8.31 Joule/deg (K) mol and F =
9.65 104 C/mol, calculate the equilibrium potential for potassium,
sodium, and chloride ions at 20 degree Celsius for the inside and
outside concentrations presented in Figure 19.1.
19.2 Calculate the reversal potential using the GoldmanHodgkinKatz
equation for the concentrations presented in Table 19.1 with the following ratios of permeability constants: pNa/pK = 0.05 and pCl /pK = 0.2.
19.3 Sketch the time course of the sodium and potassium permeabilities
using Figure 19.2 as the template. Sketch the behavior of the action
potential produced by the sodium and potassium currents over that
time period. Assume a maximum positive value of +20 mV and a
maximum negative value of -70 mV, and indicate in the plot where the
action potential will reach these maximum and minimum values.
20
Neural Rhythms
The focus in the last chapter was on the potassium and sodium ion channels responsible for generating action potentials in neurons. This subject is
developed further in the present chapter where the emphasis shifts towards
how ion channels generate rhythmic discharges in neurons. Voltage-gated
ion channels, currents owing through ligand-gated ion channels located in
chemical synapses (synaptic currents), and electrical synapses (gap junctions)all these contribute to the generation of neural rhythms. A variety
of patterns can be produced, some in small populations of cells and others
in large ones. Rhythmic patterns among large populations of neurons are
associated with different sleep states and with states of arousal and attention. Small circuits of neurons known as central pattern generators generate
rhythmic behavior in motor systems. These circuits control heartbeat,
regulate respiration, and control locomotion, chewing, and digestion.
This subject of the nervous system concludes in the last chapter, where
the focus will be on the chemical synapseon how it is organized and how
it promotes learning and memory formation. The organization of the
sensory cortices will be looked at along with how they process sensory
information that is relayed in from sensory organs. Special attention will be
given to the glutamate receptor ion channels and their central role in
sensory information processing, learning, and memory formation.
This chapter is divided into three parts. In the rst part, pacemaker cells
will be introduced in a discussion of heartbeat. In the middle part of the
chapter, the low frequency synchronous ring of large populations of cells
will be examined. The main focus will be on sleep oscillations and how these
forms of rhythmicity may change into abnormal epileptic seizures. The third
part of the chapter will deal with how central pattern generators drive
muscle contractions, drawing on several invertebrate systems for examples.
488
Description
+
Function
Fast upstroke
Diastolic depolarization
Plateau
Late repolarization
Early repolarization
489
vated by large depolarization while a few, most notably the T-type channels,
require only mild depolarization of the membrane. The calcium channels
requiring large depolarization are referred to as high voltage-activated
(HVA) calcium channels whereas the T-type channels are termed low
voltage-activated (LVA). The calcium currents found in the heart help maintain action potential plateaus, the fairly at and slowly declining portions
of the action potentials. Lastly, the potassium currents related in the last
two sets of entries in Table 20.1 are responsible for the early and late phases
of action potential repolarization.
490
Figure 20.2. Action potentials in different parts of the heart: Shown are action
potentials in the sinoatrial node (SAN) and in the ventricles. The latter are delayed
by the amount of time required for the pulse from the SAN to reach the ventricular bers. The minimum, hyperpolarized values of the membrane potentials are
listed just below the dashed lines.
Table 20.2. Collective ring patterns observed in electroencephalograph recordings in the brain.
Type of activity
Frequency
Association
Delta waves
Theta waves
Alpha waves
Spindling oscillations
Beta waves
Gamma band
0.5 to 4 Hz
4 to 8 Hz
8 to 12 Hz
7 to 14 Hz
12 to 30 Hz
30 to 60 Hz
C-terminal region. Its voltage regulation is unusual. Most ion channels are
activated by depolarization. As discussed in the last chapter, their activation gates are closed at resting membrane potentials but open when the
membrane is depolarized beyond some threshold level. The HCN channels
work the opposite way. They are activated when the membrane potential is
hyperpolarized beyond values in the range -50 to -70 mV. Whenever a
cell res an action potential the membrane hyperpolarizes and the HCN
491
492
tle back its production of cAMP. As a result the activation function shifts
to more negative membrane potentials and the heart rate slows down.
493
494
increases and then decreases back to zero, terminating the burst of action
potentials. The calcium channels that open to start the burst then require a
large hyperpolarization in order to de-inactivate and start the next burst.
Tonic ring and rhythmic bursting are widely encountered in the brain,
and neurons can switch from one form to the other. Large populations of
neurons in the brain undergo slow rhythmic bursting during natural, slow
wave sleep, but switch their ring patterns from bursting to tonic ring in
waking states and rapid eye movement (REM) sleep. In order to transition
from a slow wave sleep state to either awake or REM sleep states, the delta,
alpha, and spindling patterns have to be abolished and replaced by the high
frequency tonic ring in the beta band (Table 20.1).
The waking up and REM sleep tasks are carried out by collections of
neurons that the wake-up system comprises, the reticular formation, located
in the brain stem. Neurons in the reticular formation release a number of
neuromodulators, the most prominent of which are acetylcholine (ACh),
norepinephrine (NE), and serotonin (5-HT). These neuromodulators inuence the activities of the neurons generating brain rhythms and also inuence muscle states. In the brain, high frequency activity replaces the low
frequency waves. The membrane potential is depolarized in response to the
neuromodulators, and the T-type calcium channels responsible for bursting
are shut down.
495
tors. Similarly, glutamate and GABA each can function either as a neurotransmitter or as a neuromodulator, depending on whether it acts through
ionotropic or metabotropic (G protein-coupled) receptors. The functional
characterization of a molecule as a hormone, as a neurotransmitter, or as a
neuromodulator is determined by the nature of its receptor and what kind
of cellular response is elicited upon binding.
Recall from Chapter 12 that G protein-coupled receptors activate heterotrimeric G proteins. Neuromodulators can exert their inuences on ion
channels in two ways. In direct regulation, G protein subunits, particularly
the Gbg subunits, diffuse along the inner face of the plasma membrane and
bind to the cytoplasmic portions of ion channel subunits, thereby modifying channel conductances. Alternatively, the neuromodulators can exert
their inuences indirectly by activating second messenger systems. The
second messengers either bind the ion channels, thereby modifying their
conductances, or they activate protein kinases and protein phosphatases,
which, in turn, phosphorylate or dephosphorylate ion channel subunits,
thereby modifying their conductances. The modied states of the ion
channel subunits have lifetimes that are long compared to the time that it
takes to generate and propagate action potentials, and thus the modulatory
inuences are long-lasting ones.
496
Figure 20.5. Connexin topology and assembly into gap junctions: (a) Connexins
pass back and forth through the plasma membrane four times. Starting from the Nterminal end the transmembrane segments are designated as M1 through M4. The
N- and C-terminal portions of the polypeptide chains are situated in the cytoplasm
of the host cell. The C-terminal end is longer than the N-terminal end and may form
a loop. The two extracellular loops, E1 and E2, mediate connexin to connexin
contacts while the three cytoplasmic portionsthe N- and C-terminal ends and the
cytoplasmic loop (CL) connecting M2 to M3are sites for regulation of the pore.
(b) Six connexins arrange themselves in a symmetric fashion to form a hemipore
called a connexon. Two connexons, one from each cell, form the conduction pore,
or gap junction. These remain separated from one another by a 2- to 4-mm gap,
denoted in the gure by the pair of parallel lines.
497
Like ion channels, gap junctions are voltage-gated. Gap junctions will
open and close, and change their permeabilities, in response to alterations in
potential difference. The state of the pore is responsive to differences in
membrane potential of the two cells (transjunctional voltage differences)
and to differences in potential across plasma membranes, i.e., between the
cytoplasm and extracellular spaces (inside-outside voltage differences). Gap
junctions are also responsive to biochemical changes in the intracellular environmentfor instance, the pores will close when exposed to low pH levels.
In addition, gap junctions are subject to regulation by phosphorylation.
498
receptors that endow the neuron with intrinsic rhythmic bursting properties. In the absence of the gap junctions and the inhibitory connections, each
excitatory neuron would carry out its program of rhythmic bursting in a distinct and uncoordinated manner. When the inhibitory circuitry is present,
the cells synchronize their ring patterns. They can undergo rhythmic discharges at low frequencies and also at the higher frequencies characteristic of the beta and gamma bands of the EEG.
499
500
These neurons are organized into small neural circuits called central pattern
generators (CPGs). The CPGs may be thought of as elementary circuits,
autonomous modules built from small numbers of neurons that are used to
drive locomotion activities such as walking and swimming, breathing, and
chewing and digestion. Many of the principles governing their operation
are shared by other rhythm-producing systems such as those responsible
for sleep and alert behavior. Processes that generate rhythms, processes
such as reciprocal connections involving inhibition, and burst-promoting
voltage-gated ion channels, are common to all rhythm-producing circuits.
Motor neurons supply input to muscles that drives their contractions.
They are the last stage of neural connectivity before the muscles. Inputs
from different drivers such as the CPGs converge upon these cells. Some
of the regulatory inputs come directly to the motor neurons while others
converge upon the CPGs to regulate their ring patterns.
Invertebrates are widely used as model systems for studying central
pattern generators.The invertebrate CPGs exhibit all of the main properties
of vertebrate CPGs, yet are small enough to be well characterized at the
network and ion channel levels.The invertebrates selected as model systems
include the nudibranch Tritonia diomedea, the sea snail Aplysia californica,
the decapod crustaceans Panulirus interruptus (spiny lobster) and Cancer
borealis (rock crab), and the medicinal leech Hirudo medicinalis.
The stomatogastric ganglion (STG) of the lobster and crab is the site of
a pair of central pattern generators that supply digestive rhythms. These
crustaceans swallow their food whole. The material ingested is broken down
in the stomach through rhythmic contractions of gastric teeth and the
stomach. Several different kinds of rhythms are produced during digestion.
One of these is the gastric rhythm that controls the gastric teeth, and
another is the pyloric rhythm that regulates the food sorting and sifting
machinery. The rhythms are supplied by two CPGs. The CPG responsible
for the pyloric rhythm contains 14 neurons. Of these 13 are motor neurons
whose output drives the muscle contractions, and one is a pacemaker
neuron. The CPG responsible for gastric rhythms contains just 11 neurons.
However, its wiring and operation is more complex than that of the pyloric
circuit, and the discussion will be limited to the latter.
The pyloric circuit is presented in Figure 20.7. There are six different
kinds of neurons in the circuit. One each of these neuron types and its connectivity is shown in the gure. Connection leading to cells outside the CPG
are omitted to maintain clarity. As can be seen in the diagram the neurons
in the pyloric central pattern generator are connected to one another by
electrical synapses and by chemical synapses. The AB neuron functions as
the pacemaker neuron for the circuit. It produces bursts of action potentials at regular intervals. Electrical synapses link the AB and PD neurons
to one another. As a result the PD neuron res in synchrony with the AB
neuron, and these two neurons may be regarded jointly as the network
pacemaker. As is shown in Figure 20.7, these two neurons establish contact
with all the downstream motor neurons.
501
Figure 20.7. Crustacean pyloric circuit: The central pattern generator responsible
for pyloric rhythms consists of anterior burster (AB), pyloric dilator (PD), ventricular dilator (VD), inferior cardiac (IC), lateral pyloric (LP), and pyloric (PY)
neurons. Resistor symbols denote electrical synapses, and lines terminating in lled
circles represent inhibitory chemical synaptic connections.
Figure 20.8. Mutual inhibition: Two cells, A and B, are coupled to one another by
means of inhibitory synaptic connections. Because of this coupling the two cells
alternatively re synchronized sequences of action potentials. The dashed lines
denote the gradual depolarization of the membrane potential towards the plateau
potential and threshold for ring action potentials.
The second main feature of the network is wide usage of mutual inhibition in which pairs of cells are reciprocally connected to each other and
inhibit one another. Mutual inhibition produces alternative ring pattern
that can drive the phased contractions of opposing muscles. A stereotypic
pattern of alternating ring by a pair of cells mutually inhibiting one
another is depicted in Figure 20.8. In the pyloric circuit, the AB and PD
neurons re rst, inhibiting the ring of the downstream neurons until the
bursting is completed. The downstream neurons then re in a sequence
502
Peptides
Corticotropin-releasing factor (CRF)
Cortistatin (CST)
Opioids (dynorphins, endorphins)
Oxytocin (OT)
Somatostatin (SST)
Tachykinins (substance P, neurokinins)
Vasopressin (AVP)
503
Plateau potentials
Post-inhibitory rebound
Network
Input signals that initiate rhythms
Feedback signals that regulate
rhythms
Highly interconnected circuits that
generate rhythms
Pacemaker cells that generate
rhythms
Regulatory
Extrinsic
Intrinsic
Description
Pacemaker and other rhythm-producing
neurons express HCN ion channels that
support rhythmic bursting
Increase excitability resulting from a stable
state lying close to thresholds for action
potential generation
Small conductance, calcium-activated
potassium channels allow for adaptation of
the ring rate
Low voltage-activated (T-type) calcium
channels permit the temporary increase in
excitability
Input from sensory neurons, and from neurons
in brain stem control areas
Mechanical and chemical signals that are used
to ne tune the rhythms
Mutual inhibition and gap junctions ensure the
proper ring sequences and produce synchrony
Cells that entrain other cells in the CPG
504
505
506
ductances for the ion channels. These changes lead to alterations in phase
of one neuron relative to others within the CPG and change the number of
action potentials generated in a given time interval.
As indicated in Table 20.4, neuromodulatory signals can originate either
within a CPG or externally to it. There are many sources of external inputs
in the central nervous system. In vertebrates these sources are organized
into centers that project out to, and make contact with, neurons in most
areas of the brain. These centers are able to inuence the response properties of many circuits simultaneously. Intrinsic modulation is far more specic in its actions. In the stomatogastric ganglion, modulatory inputs are
supplied by higher brain centers and by feedback from sensory neurons.
These modulatory inputs are sensitive to the outputs of the circuit being
modied and are local to that circuit. These inuence the rhythms being
produced and the attendant behaviors.
507
Figure 20.9. Tritonia escape circuit: Upstream signals conveyed by a set of about
80 S cells are relayed to the command neuron, the dorsal ramp interneuron (DRI),
which sends its synaptic output to a set of three dorsal swim interneurons (DSIs)
belonging to the central pattern generator. The CPG contains, besides the DSIs, a
pair of ventral swim interneurons (VSIs) and a single cerebral cell 2 (C2) neuron.
The three kinds of CPG neurons make synaptic contacts with a set of about 55
dorsal and ventral exion motor neurons. In the diagram, sharp arrows denote
excitatory connections; lled circles represent inhibitory connections, and the
hybrid arrow/circle symbol denotes a multiple component (excitatory and
inhibitory) monosynaptic contact.
508
Heartbeat
Boyett MR, Honjo H, and Kodama I [2000]. The sinoatrial node, a heterogeneous
pacemaker structure. Cardiovasc. Res., 47: 658687.
Schram G, et al. [2002]. Differential distribution of cardiac ion channel expression
as a basis for regional specialization in electrical function. Circ. Res., 90: 939950.
HCN Channels
DiFrancesco D [1999]. Dual allosteric modulation of pacemaker (f) channels by
cAMP and voltage in rabbit SA node. J. Physiol., 515: 367376.
Ludwig A, et al. [1998]. A family of hyperpolarization-activated mammalian cation
channels. Nature, 393: 587591.
Zagotta WN, et al. [2003]. Structural basis for modulation and agonist specicity of
HCN pacemaker channels. Nature, 425: 200205.
Thalamocortical Rhythms
Huguenard JR, and Prince DA [1992]. A novel T-type current underlies prolonged
Ca2+ dependent burst ring in GABAergic neurons of rat thalamic reticular
nucleus. J. Neurosci., 12: 38043817.
McCormick DA, and Huguenard JR [1992]. A model of the electrophysiological
properties of thalamocortical relay neurons. J. Neurophysiol., 68: 13841400.
Munk MHJ, et al. [1996]. Role of reticular activation in the modulation of
intracortical synchronization. Science, 272: 271274.
Steriade M, Amzica F, and Contreras D [1996]. Sunchronization of fast (3040 Hz)
spontaneous cortical rhythms during brain activation. J. Neurosci., 16: 392417.
Steriade M, McCormick DA, and Sejnowski TJ [1993]. Thalamocortical oscillations
in the sleeping and awake brain. Science, 262: 679685.
von Krosigk M, Bal T, and McCormick DA [1993]. Cellular mechanisms of a
synchronized oscillation in the thalamus. Science, 261: 361364.
Epilepsy
Crunelli V, and Leresche N [2002]. Childhood absence epilepsy: genes, channels,
neurons and networks. Nature Rev. Neurosci., 3: 371382.
Velazquez JPL, and Carlen PL [2000]. Gap junctions, synchrony and seizures. Trends
Neurosci., 23: 6874.
509
Gap Junctions
Bruzzone R, White TW, and Paul DL [1996]. Connections with connexins: The
molecular basis of direct intercellular signaling. Eur. J. Biochem., 238: 127.
Evans WH, and Martin PEM [2002]. Gap junctions: Structure and function. Mol.
Mem. Biol., 19: 121136.
Kumar NM, and Gilula NB [1996]. The gap junction communication channel. Cell,
84: 381388.
Simon AM, and Goodenough DA [1998]. Diverse functions of vertebrate gap
junctions. Trends Cell Biol., 8: 477483.
Unger VM, et al. [1999]. Three-dimensional structure of a recombinant gap junction
membrane channel. Science, 283: 11761180.
Respiration
Gray PA, et al. [1999]. Modulation of the respiratory frequency by peptidergic input
to the rhythmogenic neurons in the Prebtzinger complex. Science, 286: 1566
1568.
Lieske SP, et al. [2000]. Reconguration of the neural network controlling multiple
breathing patterns: Eupnea, sighs and gasps. Nature Neurosci., 3: 600607.
Ramirez JM, and Richter DW [1996]. The neuronal mechanisms of respiratory
rhythm generation. Curr. Opin. Neurobiol., 6: 817825.
510
Problems
20.1 A small number of ion channels have particularly prominent roles in
generating rhythmic patterns of action potentials. One of these, the
HCN channel, was discussed in Section 20.2 while another, the T-type
calcium channel, was discussed in Section 20.4. Make a sketch of the
activation and deactivation functions for the T-type calcium channel
and the sodium channels responsible for action potential upstrokes.
Recall from the discussion in Section 20.4 that at the resting potential
the T-type calcium channel is activating while the sodium channel is
deactivating. In the plots of current versus membrane potential, indicate the location of a resting potential and position the four curves so
they satisfy the relationship just mentioned.
20.2 The utility of a gradually depolarizing membrane potential was discussed in Section 20.11 and illustrated in Figure 20.1. As noted in that
discussion, this behavior greatly enhances excitability in cells possessing plateau potentials. When two cells of this type are coupled through
mutual inhibition they can synchronize their ring acting as oscillatory
units that produce motor rhythms. List some types of currents that
promote gradual declining membrane potentials of the sort depicted
in the gure.
21
Learning and Memory
The learning and memory formation processes carried out by the snails and
other invertebrates is certainly more modest than that of humans, yet the
two are similar. Each involve alterations in behavioral responses to stimuli
brought on by experience. Learning is the adaptive process whereby
changes in behavior are induced in response to experience. Memory is the
record, or trace, underlying the changes in behavior. The records in all
organisms, snails and mice, ies and humans, are stored in the patterns of
connection between neurons. During learning and memory formation some
connections are strengthened while others are weakened. For instance, in
the case of sensitization discussed at the end of the last chapter, connections between neurons were strengthened. This led to behavioral changes.
The snails were able to respond with increased rapidity to noxious stimuli.
The term efciency of synaptic transmission refers to the magnitude of
the response generated in a postsynaptic neuron when an action potential
is generated in the presynaptic neuron. In many situations, the efciency of
the transmission is not xed but instead varies in ways that depend on use.
The changes in efciency can be transient, lasting for a few milliseconds, or
can be permanent, lasting for a lifetime. Connections may be strengthened
or be weakened or disappear altogether. Synaptic plasticity is the term used
to describe the use-dependent, or adaptive, changes in the efciency of
synaptic transmission between pre- and postsynaptic cells.
The ability to vary connection strengths is an important one. It allows the
nervous system to form its precise connections and neural circuits during
development; it gives rise to a unique personality for each individual; and
it makes possible learning throughout life. Cortical circuits develop over
time in several stages. The rst stage of circuit development involves neurite
outgrowth, growth cone pathnding and then synaptogenesis. As the circuits become functional and can respond to synaptic input, ineffective and
improper connections are pruned, and neurons that are no longer needed
die off through programmed cell death. The process of varying the strengths
of synaptic connections continues throughout life. It underlies the development of simple motor skills as well as highly coordinated eye and hand
511
512
513
Figure 21.2. Architecture of the brain showing the sequestering of brain functions
within specic areas of the brain: The locations of the major sensory areas are shown
along with motor areas and sites where forms of learning and memory, and higher
processing functions, as most strongly associated.
The cerebellum is a large region that sits behind the brain stem and below
the occipital lobe.Neurons in this region of the brain integrate and coordinate
motor actions and mediate motor learning. Cerebellar neurons control
balance and posture, and ensure that ne motor actions are accurate. The
large section of cortex sitting above and about these structures is partitioned
into frontal, parietal, occipital, and temporal lobes. The temporal lobe sits
above and to the outside of the brain stem at about the level of the ears. As
indicated in Figure 21.2, neurons in this region are involved in the senses of
sound,taste,and smell,and in memories thereof.Information of a tactile character, that is, the sense of touch, is processed by neurons located in the
somatosensory cortex that lies above in the parietal lobe. This region is situated next to regions where associations and interpretations are made from
information sent from the primary visual cortex and other primary sensor cortices.Behind this region,at the back of the head in the occipital lobe,(primary)
visual information is received and processed. Lastly, the nonmotor portions
of the frontal cortex are used for making judgments and intelligent decisions.
The premotor cortex controls head and eye movements, while speech is centered in Brocas area and smell in the olfactory center.
The presence of a large region devoted to motor and sensory information
processing is unique to mammals.This region is highly differentiated radially
into layers and laterally into regions. As rst noted by Brodmann in 1909,
regions of cortex involved in processing sensory information are organized in
the radial direction into six layers and in the lateral direction into at least 50
distinct areas. The numbers of sublayers, and their thicknesses and arrangements, vary from area to area in the cortex.Within each area there are cells of
a particular size and arborization, with a distinct set of inputs, outputs, and
514
515
516
517
three headings in Table 21.1. The rst set of signaling elements in the table
is the voltage-gated calcium channel grouping that converts membrane
depolarization into a calcium signal. Included under this heading are a
number of signaling proteins that allow neuromodulators to inuence the
strength of the calcium signals and thus the amount of neurotransmitter
released into the cleft.
The second grouping contains vesicle regulators such as the soluble NSF
attachment protein receptor (SNARE) complex. SNARE proteins regulate
the nal stages of neurotransmitter releasedocking and fusion of the
vesicle with the plasma membrane resulting in release of the vesicle contents into the synaptic cleft. There are two classes of SNARE proteins
those that bind to the vesicle surface (v-SNAREs) and those that attach to
the plasma membrane (t-SNAREs). Interactions between the two kinds of
SNARES regulate the release of neurotransmitter. One of the v-SNAREs,
synaptotagmin, contains a calcium-binding C2 domain and functions as the
calcium sensor. Conformational changes brought on by calcium binding are
conveyed to the t-SNAREs with which it interacts promoting fusion and
vesicle content release.
The other signaling complexes in the vesicle regulator grouping help
shepherd vesicles into the presynaptic terminal and to the plasma membrane, and then regulate membrane fusion. Neurotransmitter-lled vesicles
Table 21.1. Main components of the presynaptic terminal at CNS synapses:
AbbreviationsN-ethylmaleimide-sensitive fusion protein (NSF); soluble NSFattachment protein (SNAP); soluble NSF attachment protein receptor (SNARE);
vesicle-associated membrane protein (VAMP); CaMK/SH3/guanylate kinase
domain protein (CASK).
Presynaptic terminal component
Function
t-SNAREs
Sec6/8 complex
Sec1/Munc18
NSF, a-SNAP
Scaffolds and adapters
CASK
Mint1
Veli
518
are transported along the cytoskeleton into the presynaptic terminal. Upon
arrival at the terminal the vesicles are targeted to the plasma membrane by
a multiple subunit protein complex called Sec6/8. Another set of proteins,
Sec1/Munc18, act both as positive and as negative regulators of SNAREmediated fusion through interactions with Syntaxin. Yet another set of molecules, NSF and a-SNAP, direct the dissociation of the SNAREs permitting
their reuse.
The third grouping presented in Table 21.1 encompasses the scaffold and
anchor proteins. These proteins help organize the complexes in the presynaptic terminal. As is the case in any cell, these proteins provide attachment
sites where proteins that must work together can bind in close proximity to
one another.
Figure 21.5. The postsynaptic density: Shown as some of the types of signaling proteins that are found in the postsynaptic density at excitatory synapses in the central
nervous system. Glutamate released at the presynaptic terminal diffuses across the
synaptic cleft and binds to NMDA and AMPA receptors leading to membrane depolarization, calcium inux, alterations in AMPA receptor population, and gene
expression. Short-term modulatory effects are mediated by GPCRs such as
mGlu5R, leading to G protein subunit- and second messenger-binding to ion channels, and phosphorylation of ion channels by protein kinases.
519
Function
GluRs: Key component of active synapses
NRs: Dual voltage and ligand-gated allowing entry of
calcium into the cell
mGluRs: Signal to IP3 regulated intracellular calcium
stores
Central signaling element, involved in both short and
long term forms of synaptic plasticity
Central signaling element, involved in both short and
long term forms of synaptic plasticity
Signaling element, involved in short term forms of
synaptic plasticity
Signaling element, involved in both short and long term
forms of synaptic plasticity
Regulator of homeostasis and learning
Sequesters protein kinase A, protein kinase C, and
calcineurin near receptors and ion channels
Attaches to GluR2 and GluR3
Attaches to NR2s
Activates MAP kinase signaling
Link between PSD-95 and GKAP
Link between Homer and GKAP
Attaches to mGluRs and to Shank
tural (cytoskeleton) and regulatory (receptor, ion channel, scaffolding, signaling) proteins. The several different kinds of regulatory proteins have
been placed into three groupings in Table 21.2. The rst group contains the
receptors for neurotransmitters. Glutamate is the predominant neurotransmitter at excitatory synapses in the central nervous system. Several different kinds of glutamergic receptors are found in the PSD of excitatory
synapses in the CNS. Some glutamergic receptors are ion channels
(ionotropic) while others are G protein coupled receptors (metabotropic).
Two kinds of ionotropic glutamate receptorsthe a-amino-3-hydroxy-5methyl-4-isoxazolepropionate (AMPA) receptors and the N-methyl-daspartate (NMDA) receptorsare intimately connected with synaptic
plasticity. Rather than listing a large number of receptors in the table, only
the three main classes of glutamergic receptors have been included in the
table. These are the principal receptors involved in learning and memory
formation.
520
The second group in the table consists of several different kinds of serine/
threonine kinases and phosphatases that are central signaling elements
in the postsynaptic density. The third grouping contains connecting
and supporting agents such as GTPases functioning as molecular adapters,
and anchoring and scaffolding proteins that help organize the signaling
complexes. These signaling elements form a matrix within the PSD that
allows ion channels to cluster together in close proximity to second messenger and downstream signaling elements. The postsynaptic density is
enriched in cytoskeleton proteins, especially in actin and actin-related
cytoskeleton components.
521
Figure 21.6. Circuitry underlying the associative Aplysia gill withdrawal response:
Three different kinds of neurons form the circuitsensory neurons that detect and
respond to electrical shock (SN1) and touch (SN2), a serotonin-releasing interneuron (INT), and a motor neuron (MN).
diffuses across the synaptic cleft to the axon terminal of the touch sensory
neuron where the two sets of signals converge.
Just as there are several types of learning, there are several kinds of
memory. Short-term memory refers to memories that are labile (responsive
to changes in conditions under which they are created) and are only transiently formed. They are lost if not converted into stable memory traces.
Short-term labile memories involve celluar changes that utilize cellular
resources that are already available and are local to the synapse or synapses
undergoing modication. In contrast, long-term stable memory traces typically involve gene expression and protein synthesis leading to architectural
modications that produce the lasting changes in synaptic transmission.
This events stimulated by the release of serotonin are depicted in Figure
21.7. These include stimulation of adenylyl cyclase leading to the production of cAMP and its subsequent activation of the catalytic subunits of
protein kinase A. Protein kinase A phosphorylates the nearby potassium
channels and, as a result, currents through potassium channels are reduced.
The depolarization accompanying arrival of the action potential in the axon
terminal lasts longer. More calcium is able to enter the terminal resulting
in a greater release of neurotransmitter. Two routes are illustrated in the
gure. One of the routes leads to ion channel modication, a short-term
memory trace. The other route leads to the nucleus and results in changes
in the pattern of gene expression, producing a longer lasting memory trace.
522
Figure 21.7. Signaling in the learning pathway in Aplysia leading to short- and longterm facilitation: Serotonin stimulates an increased production of cAMP by adenylyl
cyclase through actions of GPCRs and Ga subunits.A transient facilitation of the signal
response is produced by phosphorylation of ion channels by the catalytic subunit
(PKAC) of protein kinase A.This modication alters the biophysical properties of the
ion channel. Long-term responses are elicited by sustained serotonin signaling resulting in gene transcription and morphological changes at the synaptic terminal.
523
terminal. The key biophysical property of these receptors is that they are
both voltage-dependent and glutamate-dependent thereby allowing them to
integrate signals and to evaluate associativity. These receptors work in the
following way:At the membranes resting potential, the NMDA receptor ion
channel is closedit is physically blocked by an Mg2+ ion that sits inside the
opening and blocks the pore.When the membrane is depolarized sufciently
the Mg2+ block is relieved, and once glutamate is bound, calcium ions can
pass through the channel and enter the cell. The voltage dependence is the
source of the receptors integrative property. No single synapse by itself can
depolarize the postsynaptic membrane sufciently to relive the magnesium
block. Rather, many synaptic inputs acting in close spatial and temporal
proximity to one another are needed to adequately depolarize the membrane. When depolarization and ligand binding occurs within an appropriate time window, the NMDA channel will open. Thus, the NMDA receptor
provides synapses with a way of determining whether the associativity conditions for synaptic modication have been satised or not.
One of the ways that both the efciency of synaptic transmission and
the ability to vary this property can be controlled is through the balance
between AMPA and NMDA receptors. At many postsynaptic terminals
only NMDA receptors are active during development. These synapses
remain silent at resting potentials because of the Mg2+ block, but are
capable of transmitting signals when the membrane is depolarized by one
means or another. In response to entry of calcium into the terminal through
the NMDA receptors, AMPA receptors become active. The presence of
active AMPA receptors allows the synapses to respond rapidly to glutamate
and generate stronger responses at the postsynaptic terminal to the release
of glutamate. AMPA receptors and AMPA receptor trafcking have a
prominent role in learning and memory formation and for that reason are
highlighted in Figure 21.5.
524
Figure 21.8. Hippocampal circuit in the rodent used in studies of LTP: The core
circuit includes a set of one-way connections from the entorhinal cortex (EC) to the
hippocampal dentate gyrus (DG), cornu ammonis 3 (CA3) region, CA1 region, subculum (Sub), and back to the EC and to other cortical regions. LTP is studied in the
CA3 region, where there is a dense network of recurrent connections, and in the
connections between the CA3 and CA1 neurons. Two sets of connections converge
on the CA1 neuronsthe Schaffer collaterals from the (ipsilateral) CA3 and the
commissural bers from the contralateral (contra) CA3.The names commonly given
to the various axons are shown.
in brain slices prepared from portions of the rodent hippocampus called the
CA1 and CA3 regions. The general architecture of the hippocampus
showing the locations of the CA1 and CA3 regions is presented in Figure
21.8. Two sets of connections are noted in the gurerecurrent connections
between cells in CA3, and connections from CA3 to CA1. In either circuit,
when a tetanic stimulus (i.e., a train of high frequency pulses) is delivered
to the presynaptic cell, or alternatively a series of pulses at low frequency
is delivered and the event paired with a laboratory-supplied depolarization
of the postsynaptic membrane, the efciency of synaptic transmission is
increased. The increase, or potentiation, of the synaptic efciency can be
generated in milliseconds, yet it lasts for minutes or even hours.
21.9 CREB Is the Control Point at the Terminus of the Learning Pathway
525
such as serotonin and dopamine that bind to GPCRs, which act through
heterotrimeric G proteins to stimulate ACs to produce cAMP.
The MAP kinase and PKA routes are highlighted in Figure 21.9. As
shown in this gure, calcium entry either through NMDA receptors or
through L-type calcium channels leads to the activation of a number of
serine/threonine kinasesprotein kinase A, protein kinase C, MAP kinases,
and calcium/calmodulin-dependent protein kinases II and IV (CaMKII and
CaMKIV), not all of which are shown the gure. Several different actions
can occur depending on the duration of the synaptic signaling. Short-term
actions can take place in the neighborhood of the receptors using resources
already available. If the synaptic signaling is sustained over time, long-term
changes can occur involving changes in gene expression.
526
527
Figure 21.11. Hebbs rule: (a) In response to the ring of an action potential by
cell A, cell B res an action potential. In this situation, the depolarization of the
postsynaptic membrane contributes to the ring of an action potential by cell B. The
strength of that synapse is increased. (b) Cell B does not re an action potential
when cell A does. The depolarization at the postsynaptic membrane is not effective
in eliciting action potentials. In situations of this sort the strength of the synapse is
reduced.
part in ring it, some growth process or metabolic change takes place in one
or both cells such that As efciency, as one of the cells ring B, is increased.
The rule in this form is incomplete. It does not state what happens when the
postsynaptic cell fails to re.To complete the rule one adds a subrule that says:
When a presynaptic axon of cell A repeatedly and persistently fails to excite
the postsynaptic cell B,while cell B is ring under the inuence of other presynaptic axons, metabolic changes takes place in one or both cells such that As
efciency, as one of the cells ring B, is decreased. In other words, synapses
whose activation is strongly correlated with the ring of the postsynaptic cell
are strengthened, while those synapses whose activation is poorly correlated
with the ring of the postsynaptic cell, for example, by being silent while the
cell res, are reduced in efciency.
According to the expanded form of Hebbs rule two types of changes in
the efciency of synaptic transmission can occur: the synapse can be
strengthened or it can be weakened. The efciency of synaptic transmission
increases when a synaptic connection is strengthened and decreases when
the connection is weakened. Modications in synaptic efciency depend
upon the timing of the action potentials in the pre- and postsynaptic cells.
Repetitive pre- and postsynaptic rings occurring within 10 to 50 msec of
one another are able to inuence the efciency of synaptic transmission. If
cell A res its action potential before cell B res its action potential, then
the synaptic connections between the two cells will be increased. If, on the
other hand, cell B repeatedly res before cell A, then the strength of the
synapse will decrease. The situation is intrinsically asymmetric so that there
is a preferred direction of information ow.
528
Figure 21.12. Sliding threshold model: Shown is a plot of the modication predicted
by the sliding threshold model as a function of the total activity in the postsynaptic cell. Activity levels below that of the threshold produce long-term depression, or
LTD (negative modication), while those that exceed the threshold lead to longterm potentiation, or LTP (positive modication). The threshold adapts to the
overall activity in the postsynaptic cell averaged over a suitable time interval.
529
synapse. Since the scaling affects all synapses in the same way it may be
best thought of as a form of cellular homeostasis operating in addition to
Hebbian mechanisms.
530
Figure 21.13. Circuitry involved in responses to fearful and dangerous sensory and
contextual stimuli: Abbreviations: LNLateral nucleus of the amygdala; BN
Basal nucleus; ABNAccessory basal nucleus; CNCentral nucleus. The hippocampal system responsible for relaying contextual information into the amygdala
from sensory regions consists of the perirhinal cortex, entorhinal cortex, and
hippocampus.
areas involved in mood and emotional responses and exhibit the following
set of characteristics:
the compulsion to take drugs,
loss of control in limiting intake,
entry into a negative emotional state when access to drugs is prevented,
and
relapses into addiction after periods of abstinence.
The changes induced by drug use are long-lived and involve changes at both
the cellular and molecular levels. The cells and circuits that underlie drug
addiction are located in several regions of the brain. They include cells
that express acteylcholine receptors and communicate using dopamine
(dopaminergic) and serotonin (serotonergic), and also include cells that
release glutamate (glutamergic) or GABA (GABAergic). The corresponding brain regions are associated with reward, mood, arousal, and cognition. Long-lasting changes in synaptic transmission similar to those
associated with learning and memory, but leading to addiction, take place
in these brain regions.
The sites of action of addictive drugs such as cocaine, amphetamines, and
nicotine lie deep in the brain, in regions associated with mood, emotions,
and learning. As shown in Figure 21.14, drugs act on neurons in the upper
part of the brain stem (ventral tegmental area and substantia nigra), the
limbic system (amygdala and hippocampus), and the mesolimbic system
531
Figure 21.14. Side view of the brain showing the main sites of drug action: The
regions of drug actionthe ventral tegmental area, substantia nigra, nucleus accumbens, striatum, and prefrontal cortexare shown in lighter and darker gray shades.
Areas that form the nigrostriatal system are shown in light gray and those that the
mesocorticolimbic system comprises are presented in dark gray.
532
amygdala, and the prefrontal cortex that encompasses the nonmotor portions of the frontal lobe.
The most striking feature of cells in the two systems is the prominence
of dopamine-releasing neurons. Neurons in the ventral tegmental area
and in the substantia nigra contain large numbers of dopamine-releasing
neurons. When stimulated by drugs these neurons elevate the extracellular
dopamine levels in the nucleus accumbens and striatum. Dopamine promotes reinforcement, in which behavioral responses linked to rewards
increase in frequency over time. In the drug-reward circuits, dopamine functions as the reward signal in response to the taking of drugs such as cocaine,
and also as a stimulant of reward-seeking behavior.
533
the glutamate signals. The effect of the glutamate is to prolong the release
of dopamine results in an LTP-like potentiation of synaptic transmission
and increase in duration of the reward signal. (When nAChRs found on
dopamergic neurons in the VTA bind nicotine they depolarize the membrane. The nAChRs located on presynaptic terminals enhance glutamate
release, thus stimulating further dopamine release. When NMDA receptors
are present, long-term potentiation of synaptic transmission is produced.)
Thirdly, nicotine binds to GABA-releasing cells, which for a while throttle
back the actions of dopamine-releasing cells, but desensitization turns this
off over time, further promoting addictive effects of the drug.
A denition of synaptic plasticity useful in discussions of drug addiction
is one describing it as the ability of circuits and systems to modify their
responses to a stimulus brought on by prior stimulation, either of the same
type or of an associated form. The connections between drug addiction and
synaptic plasticity have been studied most intensively in the ventral tegmental area and the nucleus accumbens. Opiates and nicotine exert their inuences through interactions with neurons of the VTA, while cocaine and
amphetamines act on neurons of the NAc. As indicated in Figure 21.15,
cocaine and amphetamines impede the ability of the dopamine transporter
(DAT) to take up dopamine from the extracellular spaces. Homeostatic regulation is lost and the dopamine levels become excessive.
Neurons in the prefrontal cortex send and receive signals from VTA and
NAc neurons, and these regions are the locus of feelings of pleasure associated with drug addiction. The networks in these areas serve as the core of
the brains reward circuitry. Again, a key element in this reward circuitry,
that is, a feature that is common to all forms of drug abuse, is the elevation
of dopamine levels leading to pleasurable sensations.
534
ing since everything turns out as expected. The dopamine release can be
regarded as a teaching signal that triggers an increased alertness whenever
something interesting or unexpected has occurred. In doing so, it strengthens connections between neurons leading to LTP and drug addiction.
Synaptic Assembly
Biederer T, et al. [2002]. SynCAM, a synaptic adhesive molecule that drives synapse
assembly. Science, 297: 15251531.
Dalva MB, et al. [2000]. EphB receptors interact with NMDA receptors and regulate excitatory synapse formation. Cell, 103: 945956.
Dean C, et al. [2003]. Neurexin mediates the assembly of presynaptic terminals.
Nature Neurosci., 7: 708716.
Penzes P, et al. [2003]. Rapid induction of dendritic spine morphogenesis by transsynaptic EphrinB-EphB receptor activation of the Rho GEF Kalirin. Neuron, 37:
263274.
Takasu MA, et al. [2002]. Modulation of NMDA receptor-dependent calcium inux
and gene expression through EphB receptors. Science, 295: 491495.
535
Impey S, Obrietan K, and Storm DR [1999]. Making new connections: Role of ERK
MAP kinase signaling in neuronal plasticity. Neuron, 23: 1114.
Kennedy MB [2000]. Signal-processing machines at the postsynaptic density.
Science, 290: 750754.
Kim JH [2003]. Presynaptic activation of silent synapses and growth of new synapses
contribute to intermediate and long-term facilitation in Aplysia.Neuron,40:151165.
Kim JH, and Huganir RL [1999]. Organization and regulation of proteins at
synapses. Curr. Opin. Cell Biol., 11: 248254.
Malinow R [2003]. AMPA receptor trafcking and long-term potentiation. Phil.
Trans. R. Soc. Lond. B, 358: 77714.
Sheng M, and Kim MJ [2002]. Postsynaptic signaling and plasticity mechanisms.
Science, 298: 776780.
Sweatt JD [2001]. The neuronal MAP kinase cascade: A biochemical signal integration system subserving synaptic plasticity and memory. J. Neurochem., 76: 110.
Thomas GM, and Huganir RL [2004]. MAPK cascade signalling and synaptic plasticity. Nature Rev. Neurosci., 5: 173182.
Ziff EB [1997]. Enlightening the postsynaptic density. Neuron, 19: 11631174.
Long-Term Potentiation
Bliss TVP, and Collingridge GL [1993]. A synaptic model of memory: Long-term
potentiation in the hippocampus. Nature, 361: 3139.
Lisman J, Schulman H, and Cline H [2002]. The molecular basis of CaMKII function in synaptic and behavioral memory. Nature Rev. Neurosci., 3: 175190.
Malenka RC, and Nicoll RA [1999]. Long-term potentiationA decade of
progress? Science, 285: 18701874.
McGaugh JL [2000]. MemoryA century of consolidation. Science, 287: 248251.
Fear Conditioning
Blair HT, et al. [2001]. Synaptic plasticity in the lateral amygdala: A cellular hypothesis for fear conditioning. Learn. Mem., 8: 229242.
McKernan MG, and Shinnick-Gallagher P [1997]. Fear conditioning incuces a lasting
potentiation of synaptic currents in vitro. Nature, 390: 607611.
Rogan MT, and LeDoux JE [1996]. Emotion: Systems, cells and synaptic plasticity.
Cell, 85: 469475.
Rogan MT, Stubli UV, and LeDoux JE [1997]. Fear conditioning induces associative long-term potentiation in amygdala. Nature, 390: 604607.
Schafe GE, et al. [2001]. Memory consolidation of Pavlovian fear conditioning: A
cellular and molecular perspective. Trends Neurosci., 24: 540546.
Shumyatsky GP, et al. [2002]. Identication of a signaling network in lateral nucleus
of the amygdala important for inhibiting memory specically related to learned
fear. Cell, 111: 905918.
536
Drug Addiction
Berke JD, and Hyman SE [2000]. Addiction, dopamine, and ther molecular mechansism of memory. Neuron, 25: 515532.
Fiorillo CD, Tobler PN, and Schultz W [2003]. Discrete coding of reward probability and uncertainty by dopamine neurons. Science, 299: 18981902.
Hyman SE, and Malenka RC [2001]. Addiction and the brain: The neurobiology of
compulsion and its persistence. Nature Rev. Neurosci., 2: 6695703.
Koob GF, Sanna PP, and Bloom FE [1998]. Neuroscience of addiction. Neuron, 21:
467476.
Mansvelder HD, and McGehee [2000]. Long-term potentiation of excitatory inputs
to brain reward areas by nicotine. Neuron, 27: 349357.
Nestler EJ [2001]. Molecular basis of long-term plasticity underlying addiction.
Nature Rev. Neurosci., 2: 119128.
Saal D, et al. [2003]. Drugs of abuse and stress trigger a common synaptic adaptation in dopamergic neurons. Neuron, 37: 577582.
Schultz W, Dayan P, and Montague PR [1997]. A neural substrate of prediction and
reward. Science, 275: 15931599.
Waelti P, Dickinson A, and Schultz W [2001]. Dopamine responses comply with basic
assumptions of formal learning theory, Nature, 412: 4348.
Problems
21.1 How does the neuron know which synapses to strengthen and which
synapses to weaken when making long-term changes involving gene
expression and protein synthesis? In a Hebbian type of rule for changing the strength of synapses, only those synapses exhibiting correlated
ring are strengthened. Referring back to Figure 21.11(a), only the
middle synapse onto cell B is strengthened. The middle synapse
must be tagged in some fashion or other, so that the results of gene
expression can be applied to that synapse alone. How might that be
done?
21.2 In a Hebbian type of rule the postsynaptic cell res an action potential. This is a cell-wide signal that a set of associations has been made.
This may be viewed as a strong form of Hebbs rule. The weak form
of the rule would only require postsynaptic membrane depolarization
by some amount. In a weaker form of Hebbian rule, an action potential is not required, but rather the convergent signals produce a depolarization. How might action potential information get conveyed to
the dendrites to elicit an immediate strengthening of the synapse?
21.3 Several different experimental protocols are utilized in the hippocampal slice preparations. All of these involve pairing of spikes in
the pre- and postsynaptic cells. One of the most effective protocols is
that of tetanic stimulations. In the last chapter, at least four different
kinds of ring modes were discussed. Which of the different modes of
Problems
537
Glossary
540
Glossary
Antagonist A molecule that binds a receptor, but the receptor does not
transmit a signal in response to the binding event. Drugs that bind in an
antagonistic fashion are known as blockers.
Antibiotic Biomolecules synthesized by fungi and bacteria that kill competing microbes.
Antibodies Receptors synthesized by B cells that recognize and bind
antigens.
Antigen (antibody generator) Foreign substance, derived from a pathogen
and expressed either on the outer surface of the pathogen or on the surface
of an antigen-presenting cell, that triggers the production of antibodies.
Antigenic variation Systematic alterations in the antigens expressed on
the outer surface of a pathogen.
Apoptosis Programmed cell death, in which there is an orderly disassembly of the cell that avoids harming neighboring cells. Also called cell suicide.
Apoptosome The major control point for converting internal stress
signals into apoptotic responses. It is located just outside the mitochondria
and is activated by the release of cytochrome c.
Associative learning Changes in the behavioral response to the weak stimulus that has been paired with a strong (positive or negative) stimulus.
Autocrine A signaling mode in which hormones secreted from a cell act
back on the cell releasing them.
Auxiliary spice sites Sites where splicing regulators bind. Auxiliary sites
located within exons are called exonic splice enhancer (ESE) and exonic
splice inhibitory (ESI) sites, depending on which regulatory outcome is supported. Similarly, intronic sites are termed intronic splice enhancer (ISE)
and intronic splice inhibitory (ISI) sites.
Bacteriophage Virus that infects bacteria; also called a phage.
Biolm A bacterial colony formed on exposed surfaces and exhibiting
cooperative behavior between members.
Branching morphogenesis Growth, invasion, and proliferation of cells
that form branched tubular structures that carry uids in the vasculature,
lungs, kidneys, and mammary glands.
Caspases Proteolytic enzymes that catalyze the cleavage of specic molecules in response to apoptosis signals.
Catch bond A bond that is strengthened by the external forces. The forcedriven enhancements in the lifetime of these bonds allow leukoctytes to be
captured by the walls of the blood vessels and begin rolling.
Glossary
541
542
Glossary
Cytokines Small signaling proteins synthesized and secreted by leukocytes, most commonly, macrophages and T cells. They convey a variety of
instructions to leukocytes and to other cells such as neurons.
Death-inducing signaling complex (DISC) The name given to the control
point responsible for converting external death signals into apoptotic
responses. It is organized by death receptors at and just below the plasma
membrane.
Denatured state The ensemble of states that a newly synthesized protein,
or an unfolded protein, populates.
Dephosphorylation The removal catalyzed by protein phosphatases of
phosphoryl groups previously added to amino acid side chain hydroxyls on
protein substrates by protein kinases.
Desensitization The process whereby a G protein-coupled receptor, or
any other receptor, loses its responsiveness to binding by its ligand.
Diffraction Scattering of light by atoms, molecules, and larger objects
resulting in departures from rectilinear motion other than reection or
refraction.
Diffusion Thermally driven movement of particles in a uid from one
locale to another produced by random collisions of the particles with the
molecules of the uid.
Distal sites DNA regulatory regions where long-range interactions
between regulatory proteins and the basal transcription machinery take
place.They may be located upstream of the core promoter,downstream of the
core promoter,in between coding regions,and inside introns.Positively acting
transcription factors that bind at these sites are called enhancers, while negatively acting transcription factors are referred to as silencers.
Domain fold Stable arrangements of multiple secondary structure elements and of two or more structural motifs into independent folding units.
Efciency of synaptic transmission Magnitude of the response generated
in a postsynaptic neuron when an action potential is generated in the presynaptic neuron.
Electrostatic complementarity The matching of hydrophobic patches, the
complementary pairing of hydrogen bond donors and acceptors, and the
matching of positive and negative charges of basic and acidic polar residues
from one surface to the other of the interface.
Endocrine A signaling mode in which hormones are secreted into the
bloodstream and other bodily uids by specialized cells and travel large distances to reach multiple target cells.
Glossary
543
Endocytosis The process whereby plasma membrane proteins and materials captured at the cell surface are packaged into vesicles and shipped to
digestive compartments for processing and recycling.
Energy landscape A graphical depiction of how the number of states
available to a protein at each value of the potential energy varies as a function of a few signicant degrees of freedom.
Envelope Lipid/carbohydrate membrane derived from the host cell that
surrounds a viral capsid.
Euchromatin Transcriptionally active chromatin with an open shape that
permits transcription factors and the basal transcription machinery to
access promoters.
Exocytosis The packaging and shipping of newly synthesized proteins destined for export and use in the plasma membrane in vacuoles that move
over the rail system and fuse with membranes at their destination.
Exons Short coding sequences separated from one another by introns in
pre-messenger RNAs.
Focal adhesions Points of contact and adhesion between the cell and the
supporting extracellular matrix. They serve as control points where growth
and adhesion signals are integrated together to govern the overall growth
and movement of the cell.
Folding funnel The shape of the potential energy landscape that arises
because there are many high energy states and few low energy ones.
Gate The part of the ion channel that opens and closes the pore through
which ions pass.
GDP dissociation inhibitors (GDIs) Enzymes bind and maintain pools of
inactive GTPases by inhibiting the dissociation of GDP from the GDPase.
Glycosylation A common posttranslational modication to proteins destined for insertion in the plasma membrane in which covalently linked
oligosaccharides that extend out from their extracellular side are added.
The modied proteins are referred to as glycoproteins.
Glycosyl phosphatidylinositol (GPI) anchors Posttranslational modications to proteins that allow them to attach to the outer, or exoplasmic, leaet
of the plasma membrane. GPI anchors are made from a complex sugar plus
a phosphatidylinositol grouping.
Growth cones Sensory structures located at the tip of advancing axonal
and dendritic processes sent out from neurites. They explore, interact
with, interpret, and respond to signaling molecules in their local microenvironment.
544
Glossary
Glossary
545
546
Glossary
Glossary
547
548
Glossary
Plateau potentials Stable membrane potentials occurring at depolarizations greater than that of the resting membrane potential. When a membrane is at a plateau potential it is far more excitable and can repeatedly
re action potentials even in the absence of sustained excitatory input from
synapses.
Platelets Cytoplasmic fragments of bone marrow cells called megakaryocytes that form clots that block blood ow at sites of injury.
Pleiotropic Multifunctional.
Polypeptide hormones Small compact polypeptide growth factors that
bind to receptor tyrosine kinases.
Pores Membrane-spanning proteins found in the outer membrane of
Gram-negative bacteria, mitochondria, and chloroplasts forming channels
that enable hydrophilic molecules smaller than about 600 Da to pass
through.
Post-inhibitory rebound A strong hyperpolarization that is quickly terminated and followed by a rapid depolarization leading to the ring of an
action potential.
Postsynaptic density Region of the postsynaptic dendrite terminal containing the machinery for neurotransmitter signal transduction.
Pre-initiation complex (PIC) Also known as the basal transcription
machinery the PIC consists of RNA polymerase II and a set of general transcription factors.
Pre-messenger RNA (pre-mRNA) Eukaryotic RNA molecule produced
by transcription from DNA. It contains exons, introns, and regulatory
sequences that provide binding sites for the splicing machinery and
regulatory proteins.
Primary splice sites Consist of (i) the 5 splice site characterized by the
presence of a binding sequence containing a guanine-uracil (GU) pair
within a longer GURAGU-like sequence, where R is a purine; (ii) the
branch site characterized by the nucleotide sequence YNYURAY, where Y
is a pyrimidine; (iii) the pyrimidine tract, a string of pyrimidine nucleotides;
and (iv) the 3 splice site characterized by either a CAG sequence or a UAG
sequence.
Primary structure The proteins covalent structure, the linear sequence of
amino acids linked to one another by peptide bond plus all disulde bonds
formed during folding.
Promoter Transcriptional regulatory region of DNA containing binding
and start sites for RNA polymerase II and binding sites for the transcription control elements.
Glossary
549
550
Glossary
Regulon Sets of operons located at well-separated loci along the chromosome-encoding proteins involved in a common physiological response.
Response regulator In a two-component system, this unit functions as
the receiver of the transferred phosphoryl group and as an output unit for
the signal. Most output response regulators function as transcription
factors.
Responsive elements Transcriptional control points, consisting of
short DNA sequences located in promoters, where transcription factors
come together and bind in a sequence-specic manner to regulate
transcription.
Reversal potential The value of the membrane potential for a particular
ion species that exactly cancels out the ow of those ions through the membrane arising from concentration differences in that ion.
Rhythmic bursting Multiple sequences of action potentials in which each
sequence, or burst, consists of a train of closely spaced action potentials separated by large interbust intervals.
Robustness A property of a system with respect to one or more of its
parameters in which feedback damps out the effect of variations in the
value of the parameter(s) on the performance of the system.
Scaffold proteins Nonenzymatic proteins that enable signaling proteins
that must work together to attach in close proximity to one another.
Secondary structure The ensemble of short segments of the polypeptide
chain that fold into a geometrically regular, repeating structure, such
as alpha helices and beta sheets, that are stabilized by networks of
hydrogen bonds.
Second messengers Signaling intermediaries that tie together events
taking place at and just below the plasma membrane subsequent to ligand
binding. Acting as coactivators and allosteric regulators they help to recruit
and organize the proteins that function as receptors and intracellular signal
transducers.
Selectivity The ability of an ion channel to pass some ions through while
preventing passage of other ions.
Selectivity lter The part of the ion channel that selects which ions are
able to pass through the pore.
Senescence A nondividing stage of cellular life entered into when a cells
telomeres become critically shortened.
Sensitization Strengthening of a behavioral response to a noxious stimulus through repeated exposures to that stimulus.
Glossary
551
552
Glossary
Index
554
Index
Index
Apoptotic proteins, mutated in cancer,
333
Arabidopsis response regulators,
152154, 153f
Arabidopsis thaliana
hormonal signaling, 152154, 152t,
153f
light sensing, 154156, 155f
Arachidonic acid, 294, 294f
ARF, 351, 351f, 354
Arf GTPase, 263t, 269270
Arginine-serine-rich domain, 403
Arrhenius formula, 108109
Aspirin, 294
Associative learning, 520, 540
Ataxia telangiectasia mutated, 120t,
333t, 342, 342t, 343f, 344,
345346, 352
ATM and Rad3-related, 120t, 342, 342t,
345346, 352
Atomic force microscopy, 232233
Atomic radii, 7374
Atrioventricular node, 488
Attention-decit hyperactivity
disorder, 291
Auditory brain area, 513, 513f
Autocrine signaling, 248, 540
Autoinducers, 428, 429f
Auxiliary splice sites, 540
Auxiliary splicing factors, 403
Auxiliary splicing sites, 403404, 403f, 540
Bacillus subtilis, 412, 416, 427t; see also
Sporulation
Bacteria, in the ecosystem, 411
Bacterial chemotaxis, see Chemotaxis
Bacterial communities, 426428, 427t
Bacterial receptors
chemotactic pathway, 150152, 150f,
150t
clustering, 148149
structure, 147, 147f
Bacteriophage lambda
decision circuit, 458459, 458f
genome, 457459, 457f, 458f
lambda counterrepressor, Cro, 458
lambda repressor, CI, 458
lifecycle, 457, 546
structure, 457, 457f
555
556
Index
Index
Caspase 9, 371, 373, 373f, 374, 375, 376,
376f
Caspase-activated deoxyribonuclease
protein, 367, 368f
Caspase recruitment domain, 257,
362363, 363t, 373, 373f
Caspases, 360363, 361f, 362f, 363t,
365366, 540
Cassettes, 419
Catabolite activator protein, see Lac
operon
Catch bonds, 540; see also Bond action
during leukocyte rolling
Catecholamines, 286
Catenins, 227
Caulobacter crescentus, 412; see also
Differentiation, in C. crescentus
Caveolae, 164, 541
Caveolins, 164
CD4, 8 receptors, see T cells
Cell adhesion, 541
Cell adhesion molecules, see Cell
adhesion receptors
Cell adhesion receptors, see Cadherins,
IgCAMs, Integrins, Selectins
and growth factor receptors in
cancer, 337338
as long glycoproteins, 221224
in immunological synapses,
212213
Cell cycle
checkpoints, 333, 346347
control system, 347, 348f, 348t
phases of, 2324
progression, 346347
Cell fate, 17, 541
Cell lysis, see Lysis
Cell polarity, 541
Cell regulation, 12
Cell wall, 2
Central pattern generators, 499500,
541
and motor learning, 506507
common features, 502504
in human respiratory system,
504505
in invertebrate motor systems,
500502
neuromodulation of, 505506
557
558
Index
Index
Denatured state, 95, 9698, 96t, 97f, 98f,
104105, 104f, 542
Dendritic cells, 188189, 189t
Deoxyadenosine triphosphate, 372,
372t, 373
Deoxyribonucleic acid, 5, 11, 14
Dephosphorylation, 542; see also
Protein phosphatases
Desensitization, 542
Detailed balance, see Principle of
detailed balance
Developmental pathways, 305306
Diacylglycerol, 122, 165166, 166f
Diastolic depolarization, 488, 488t, 490f
Differentiation, in C. crescentus,
424426, 425f
Diffraction, 542; see also Braggs law,
X-ray crystallography
Diffusion, 134136, 138, 185186, 542
Diffusion coefcient, 135136, 138
Dipole forces, see Macromolecular
forces
Discosoma striata, 57
Dishevelled adapter, 315, 315f, 316f
Disorder, see Entropy
Distal control region, 387389, 388f,
542
Disulde bridges, 73t, 7475
DNA-binding domains, see
Transcription factors
DNA damage, 331, 340342, 341t
DNA fragmentation factor, 367
DNA microarrays, 45, 46t, 6263
DNA-PK, 120t, 342t, 345346, 352, 354
Domain fold, 542; see also Domains
Domains, 29, 3032, 31f, 32t
Dopamine, 286, 290t
Dopamine GPCR, 277t, 291
Dopaminergic neurons, 532533, 532f
Dopaminergic system, 291292
Dopamine transporter, 532f, 533
Double-strand break repair, 341, 341t,
342346
Downstream promoter element,
387389, 388f
Drosophila, 305306, 321327
Drug addiction, 291, 529530
aberrant synaptic plasticity,
532533
559
characteristics, 530
reward circuitry, 531532, 532f
sites of action, 530531
Drugs, and G protein-coupled
receptors, 16
Dual specicity kinases, see Mitogenactivated protein kinase
modules
Dynorphins, 290, 290t
E1A, 351352, 351f
E2Fs, 348, 350f, 351352, 351ff, 354
Early repolarization, 488489, 488t,
489f
Ecdysone, 393t
Ectodomain shedding, 311
Efciency of synaptic transmission,
511, 526527, 527f, 542
EF hand domain, 32t
EF-Tu elongation factor, 263, 266
EGF-like domain, 32t
eIF4E binding proteins, see Eukaryotic
initiation factors
Electrical synapses, see Gap junctions
Electroencephalographic recordings,
492
Electromagnetic radiation, interactions
with matter, 4649, 48f; see also
Braggs law, X-ray
crystallography
Electromagnetic spectrum, 47, 47f
Electron crystallography, 53
Electrostatic complementarity, 78, 542
Emission, of photons, see
Electromagnetic radiation
Endocrine glands, 247
Endocrine pancreas, 285, 287288, 287t
Endocrine signaling, 248, 542
Endocytosis, 9, 268269, 543
Endonuclease G, 372, 372t
Endoperoxide H synthases, 294
Endoplasmic reticulum, 3t, 67, 8f
Endorphins, 290, 290t
Endosomes, 3t, 9
Endpoints, of signaling pathways, see
Control point
Energy barrier
kinetic shift, 105
thermodynamic shift, 105
560
Index
Index
Fluorescence, 49, 5759, 60f
Fluorescence recovery following
photobleaching, 138
Fluorescence resonance energy
transfer, 46, 46t, 5859, 60f, 67
Fluorescent proteins, 5758, 58t, 138
Fluorophores, 49
Focal adhesion kinase, 258t, 261262,
262f
Focal adhesions, 260262, 543
Focal adhesion targeting sequence, 262,
262f
Folding cage, 102103, 103f
Folding diseases, see Misfolded
proteins and diseases
Folding funnel, 543; see also Protein
folding
Follicle-stimulating hormone, 286, 287t
Forkhead-associated domain, 255256,
257t
Forkhead transcription factors,
176177, 176f
Four-helix bundle, 29f
Free radicals, 371
Friction coefcient, 135136
Frizzled, 315, 315f
Frontal lobe, 512f, 513, 513f
Fruiting body, 434
Frustration, 107108
Functional epitopes, in binding,
202203
Funny (f) channels, see HCN channels,
voltage-gated
Fused genes, 340
Fused protein, 318, 319f
FYVE domain, 169170, 170t
GABA, 495
GABAA and GABAC receptors, 497
subunits and assembly, 499
GABAB receptors, 277t, 288, 288t, 497
GABA-releasing neurons, 497
in epilepsy, 498499
in spindling, 498
synchronization by, 497
Gamma rays, 47f
Gamma-secretase, see Secretases
Gamma waves, 490t, 492
Gap genes, 321322, 321t, 323324, 324f
561
562
Index
Index
G a subunits, 279281, 280t, 281t,
282f
G bg subunits, 279, 281t, 283, 284,
284f, 285f, 289
in protein kinase C signaling,
179180, 180f
in the cAMP signaling pathway,
180182, 181f
in yeast general stress response, 130,
130f
in yeast pheromone response
pathway, 125128, 126f
regulation of ion channels, 297
High osmolarity glycerol pathway,
128129, 128f
Hippocampus, 512, 512f, 513f, 523,
530531, 531f; see also
Long-term potentiation
architecture and circuitry, 524, 524f
Hirudo medicinalis, 500
His-Asp phosphotransfer, 140141,
141f
Histamine, 290, 290t, 293, 293t
Histamine GPCR, 277t
Histidine kinases, 141145; see also
CheA histidine kinase
Histidine phosphotransfer protein, see
Phosphorelay
Histone acetyltransferases, 41, 395396
Histone deacetylases, 41, 395396
Histone modication, 14, 386, 386t,
395397
Histones, 6, 2223
posttranslational modications of,
4041, 41f
hMLH1, mutated in cancer, 333t, 341
hMSH2, mutated in cancer, 333t, 341
HodgkinHuxley equation, 470471
equivalent circuit, 471472, 472f
h, m, n gates, 472474
Holoenzyme, 544
Homeobox domain, 32t
Homeostasis, 11, 544
Homologous recombination, 341, 341t,
342344, 342t, 343f
Homology boxes, in CheA, see CheA
histidine kinase
Hookes law, see Macromolecular
forces
563
564
Index
Index
LennardJones, 612 potential, 8183,
83f
Lentivirus family, 445t
Leucine-rich repeat, 32t
Leucine zippers, 390391, 391f
Leukocytes, 187, 188189, 189t,
190192, 230234, 243245,
545
Ligands, 16, 113
Light sensing, 154157, 154t, 155f
Lipid-binding domains, 169170,
170t
Lipid kinases, 165; see also
Phosphoinositide-3-OH kinase
Lipid phosphatases, see PTEN lipid
phosphatase
Lipid raft, 164, 545
Lipid second messengers, 122
Lipopolysaccharides, 187188, 198
Localization, see Compartmentalization
London forces, see Macromolecular
forces
Long-term facilitation, 520, 545
Long terminal repeats, 449450, 450f
Long-term memory, 521
Long-term potentiation, 512, 523524,
524f, 525526, 525f, 526f, 545546
Lou Gehrigs disease, 100, 100t, 360
Low-voltage-activated calcium
channels, see Calcium channels
LRP co-receptor, 315, 315f
Luminescent bacteria, 427428, 427t
Lutenizing hormone, 286, 287, 287t
Lymphocytes, 188189, 189t
Lysis, 443
Lysogenic cycle, see Bacteriophage
lambda
Lysosomes, 3t, 8, 8f
Lytic cycle, see Bacteriophage lambda
Macromolecular forces, 71, 7476, 76t,
7983, 81f, 82ff, 83f
Macrophages, 189t
Mad cow disease, 100
MAD homology domains, 313314
Magnetic dipole moments, 56
Magnetic resonance imaging, 57
Major histocompatability complexes,
207209, 213
565
566
Index
Index
Nicotinic acetylcholine receptors, see
Acetylcholine receptors
Nijmegen breakage syndrome, 342
Nijmegen breakage syndrome proteins,
333t, 342, 342t, 346, 354
Nitrogen xation
by cyanobacteria, 435436
by rhizobia, 436437
NK cells, 189t
NMDA receptors, 515, 519, 519t
and AMPA receptors, 523
biophysical properties, see Glutamate
receptors
dual voltage-ligand gated, 522523
in hippocampal LTP, 525, 525f
Nociception, 292293
Nod factors, 436437
Noncovalent bonds, 7576
Nonhomologous end-joining, 341, 341t,
342t, 344
Nonpolar side chains, 7374, 73t
Nonreceptor tyrosine kinases, 247,
258262, 258t
Nonsteriodal anti-inammatory drugs,
293295
Nonsteroid lipophilic hormones, 393,
393t
Noradrenaline, 286287, 287t, 290t
Norepinephrine, 494
Notch pathway, 3637, 306311, 306t,
307f, 308f, 310f
intracellular domain, 307, 308f
lateral inhibition, 307308, 309f, 545
Nuclear hormone receptors, 393, 393t
Nuclear import and export, 14
Nuclear magnetic resonance, 45, 46t,
5357, 54f, 55f, 68, 68f
Nuclear magneton, 56, 6768
Nuclear pore complex, 6, 267
passage by viruses, 442443
Nuclear spin, 54
Nucleocapsid, 546
Nucleocytoplasmic shuttling, 125128
Nucleoid region, 2
Nucleoli, 25
Nucleophile, 144, 158
Nucleosome, 22, 23, 23f, 24f, 547
Nucleotide excision repair, 340341,
341t
567
568
Index
Index
Platelets, 230, 548
Pleckstrin homology domains, 32t,
169170, 170t, 174
in phosphoinositide-dependent
kinase-1, 177f
in phospholipase C, 166, 167f
in protein kinase B, 174175, 177f
Pleiotropic, 13, 190, 548
Plexins, 236t, 239
Polar side chains, 7374, 73t
Polyacrylamide gel electrophoresis, see
Gel electrophoresis
Polyadenylate tail or poly (A) tail,
404405
Polymerase chain reaction, 62
Polypeptide hormones, 247248, 548
Polyproline (PP II) helix, 256
Polypyrimidine tract, see Pre-mRNAs
Pons, 512, 512f
Pores, 4, 548
Porins, see Pores
Post inhibitory rebound, 503t, 504,
548
Postsynaptic density, 514, 518520, 518f,
519t, 548
Postsynaptic terminal, see Postsynaptic
density
Posttranslational modications, 13, 21
anchors, 33, 34, 34t, 35ff
glycosylation, 33, 36
proteolysis, 33, 3637
to histones, 4041, 41f
to side chains, 33, 3640, 38ff
Potassium channels, voltage-gated, 475t
chain topology, 476f
conducting pore, 476477, 479, 480f
selectivity lter, 476477, 478479
subunit assembly, 476477
voltage sensor, 478479, 479f
Potassium currents, 466468, 466f, 468t,
468470, 469f; see also
HodgkinHuxley equations
Potential energy, of a macromolecule,
7983
Poxvirus family, 445t, 446t, 447
pRb, see Retinoblastoma protein
Prebtzinger complex, 504
Predictive learning, 533
Prefrontal cortex, 531f, 533
569
570
Index
Quantum mechanics, 84
Quaternary structure, 549; see also
Protein structure
Quorum sensing, 428430, 429ff, 549
Rab GTPase, 263t, 268269, 269f
Radio waves, 47f
Rad proteins, 342344, 342t, 343f
Raf, 253f
Ramachandran plot, 27, 28f
Raman spectroscopy, 61
Random coils, 28
Ran GTPase, 263t, 267268, 268f
Raphe nuclei, 292
Rapid eye movement (REM) sleep,
492, 494
Ras-binding domain, in PI3 kinase,
168f
Ras GTPase, 253f, 263266, 263t, 265f
as activators of MAP kinase
modules, 196, 197f
in cell-cycle control, 351, 351f
in T cell receptor signaling, 210f
in yeast general stress response, 130,
130f
mutated in cancer, 333t, 334336,
335ff
Reactive oxygen species, 331, 341,
371372, 375376, 376f, 378, 549
Receiver domain, 140141, 141f
Receptor clustering, 148149
Receptor for activated C-kinase,
179180, 180f
Receptor interacting proteins, 368, 368f
Receptors, 12, 112113, 112f, 549
Receptor tyrosine kinases, 247,
252254, 253f
Reciprocal inhibition, see Mutual
inhibition
Recruitment, 114, 161
Rectifying, 549
Regulator-of-G-protein-signaling
domain, 256, 257t
Regulators of G protein signaling, 279
Regulons, 419, 550
Repair proteins, mutated in cancer,
333, 333t
Reporter protein, 59, 60f
Respiration, 504505
Index
Response regulators, 140141, 141f,
145146, 145f, 145t, 550
Responsive elements, 118, 387389,
388f, 550
Resting potential, 468
Restriction point, 346f, 347
Reticular formation, 494, 512, 512f
Retinal photoisomerization, 295296,
296f
Retinitis pigmentosa, 99
Retinoblastoma protein, 333t, 347349,
350352, 350f, 351ff, 354
Retrograde signals, 273
Retroviruses, 336, 441, 449450, 450f;
see also Human
immunodeciency virus
Reversal potential, 467, 550
Reward-driven behavior, 533534
Rhabdovirus family, 445t
Rheovirus family, 445t, 446t
Rhizobia, 436437
Rhodopsin GPCR, 154t, 277, 277t,
278279, 278f, 295296, 296f
Rho GTPase, 253f, 263t, 265f, 266267
as activators of MAP kinase
modules, 196, 197f
in the Wnt pathway, 316f, 317
Rhythmic bursting, 492494, 493f, 550
Ribonucleoprotein complexes, 401403,
402f
Ribosomal S6 kinase, 120t, 133, 133f
Ribosomes, 7, 131
RING nger, 32t, 374375, 374f
RNAP holoenzyme, bacterial, 412413,
413t, 413f, 414t, 418; see also
Sigma factors
RNA polymerase II, 385, 386387,
387f, 393, 394396, 394t, 395f,
396f
RNA recognition motifs, 404, 404f
Robustness, in the chemotactic
pathway, 150152, 150f,
550
Rossman fold, 29
Rouleaux, 231
Roundabout, 238, 238f
Roux sarcoma virus, 336
R-Smad, see Smad proteins
Ryanodine receptor, 377
571
572
Index
Index
Supersecondary structures, 29
Suppressor of Fused, 318, 319f
Suppressor of Hairless, 306t, 307, 308f
SWI/SNF complex, 386, 386t, 392393,
398t
Synapse, 213, 234235, 514515, 515f;
see also Postsynaptic density,
Presynaptic terminal
Synaptic cleft, 514515, 515f
Synaptic current, in HodgkinHuxley
equation, 471
Synaptic plasticity, 511, 526527, 527f,
528529, 528f, 536, 551
in drug addiction, 533
Synaptic signaling, 248249, 551
SynCAMs, 514515, 515f
T1, T2 cells, 191t, 192
TACE metalloprotease, 311
Tachykinins, 290
Tap42 protein, see Protein phosphatase
1/2A
Targets of rapamycin proteins, 120t,
131
Targets of rapamycin signaling,
131134, 132f, 133f, 134f
Tar receptor, see Bacterial receptors
TATA-binding protein, 386387, 387f,
394, 394t
TATA box, 386387, 387f
TBP-associated factors, 394, 394t
T cells, 189, 189t
CD4, CD8 co-receptors, 209210,
210f
cytokines, see Interleukins
differentiation, 192, 192f
receptors, 207212, 210f, 211t
Telomerase production, 354355
Telomere, 354355, 551
Temporal lobe, 512f, 513
Terminal oligopyrimidine tract, 133,
133f
Tertiary structure, 552; see also Protein
structure
TFIIA structure, 391, 392f
TFIIB-recognition element, 386,
387f
TGF-b pathway, 311314, 312f
in cancer, 338339
573
574
Index