Download GEFs: master regulators of G

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

LSm wikipedia , lookup

Thylakoid wikipedia , lookup

Silencer (genetics) wikipedia , lookup

Gene expression wikipedia , lookup

Biochemistry wikipedia , lookup

Transcriptional regulation wikipedia , lookup

Histone acetylation and deacetylation wikipedia , lookup

Cell-penetrating peptide wikipedia , lookup

SNARE (protein) wikipedia , lookup

Proteasome wikipedia , lookup

Gene regulatory network wikipedia , lookup

Endomembrane system wikipedia , lookup

Magnesium transporter wikipedia , lookup

Interactome wikipedia , lookup

Nuclear magnetic resonance spectroscopy of proteins wikipedia , lookup

Biochemical cascade wikipedia , lookup

Protein moonlighting wikipedia , lookup

Protein wikipedia , lookup

SR protein wikipedia , lookup

QPNC-PAGE wikipedia , lookup

Protein adsorption wikipedia , lookup

Cyclol wikipedia , lookup

Paracrine signalling wikipedia , lookup

Western blot wikipedia , lookup

List of types of proteins wikipedia , lookup

Intrinsically disordered proteins wikipedia , lookup

Protein–protein interaction wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

Transcript
266
Forum
TRENDS in Biochemical Sciences Vol.26 No.4 April 2001
Q&A
GEFs: master regulators
of G-protein activation
What is a GEF?
GEF stands for guanine-nucleotide
exchange factor, any of a large and diverse
group of proteins that catalyze the release of
GDP and the subsequent uptake of GTP by
members of the Ras and heterotrimeric
G-protein superfamilies (referred to here
collectively as G proteins). When bound to
GTP, G proteins are in the active
conformation. Upon hydrolysis of this bound
GTP, by virtue of their own intrinsic GTPase
activities, G proteins are inactivated. GDP
release is the rate-limiting step for the
GTPase reaction in vitro; in vivo, this kinetic
barrier is reduced by GEFs. Certain GEFs
have also been called guanine-nucleotide
release factors (GRFs) or guanine-nucleotide
dissociation factors (GDFs). GEFs are not to
be confused with guanine-nucleotide
dissociation inhibitors (GDIs), which trap G
proteins in an inactive GDP-bound state, nor
with GTPase activating proteins (GAPs),
which accelerate the rate of GTP hydrolysis
(i.e. augment the intrinsic GTPase activity
of the G protein) and thereby terminate
G-protein signaling. GEFs constitute a
highly diverse group of proteins in the cell.
All GEFs contain one or more G-protein
interaction domain that manifests exchange
activity; most also contain other proteininteraction and regulatory domains.
control for most G-protein-regulated
pathways. The array of cellular activities
regulated by GEFs reflects the functional
diversity of G proteins themselves; hence,
various GEFs are involved in
transcriptional regulation (by modulating
the activity of Ras, Rap and Ral G proteins),
cytoskeletal rearrangement and motility
(via Rho, Rac and Cdc42), vesicle trafficking
(via Rab and ARF) and nuclear import (via
Ran). A survey using the LocusLink utility
of the NCBI (National Center for
Biotechnology Information) server
(http://www.ncbi.nlm.nih.gov/LocusLink/)
reveals the existence of nearly 80 genes
encoding small protein-directed GEF
homology domains in the human genome.
Heptahelical G-protein-coupled receptors
(GPCRs) constitute an enormous family of
ligand-stimulated GEFs that activate
heterotrimeric G proteins. In addition,
exchange factors IEF1B and EFTs act upon
IEF1A and EFTu, respectively, to ensure
the fidelity of the initiation and
translocation steps of ribosomal protein
synthesis. GEFs that target signaling
G proteins respond to extra- or intracellular
signals, which can induce translocation of
the GEFs to specific membrane
compartments in the cell where they then
activate G proteins. Certain GEFs also
mediate crosstalk between different
receptor-mediated signaling pathways,
thereby acting as signal integrators.
Are there many varieties of GEF?
Why are GEFs important?
By catalyzing the release of GDP from,
and the subsequent uptake of GTP by,
G proteins, GEFs enable G proteins
to activate downstream effectors such
as kinases, adenylate cyclases and ion
channels. GEFs serve as the initial point of
Members of each major subdivision of the
Ras G-protein superfamily are recognized
by one or more homologs of a distinct and
corresponding family of GEFs. For
example, the Rho–Cdc42–Rac-family
G proteins are substrates of GEFs that
contain tandem Dbl homology (DH) and
Fig. 1. A gallery of GEFs, with Protein Data Bank (http://www.rcsb.org/pdb/) accession
numbers in parentheses. (a) Elongation factor Ts from Thermus thermophilus (1IAP);
pleckstrin homology (PH) domains1. GEFs
that act upon Ras, Ral and Rap contain
domains homologous to that in the cdc25
protein of Saccharomyces cerevisiae2. A
separate class of sec7-type GEFs couple
with the ARF-family of G proteins that are
involved in vesicular transport3. GEFs
with similar guanine-nucleotide exchange
domains can act on different G proteins of
the same class, contain different sets of
regulatory and localization domains, or
exhibit different patterns of tissue-specific
expression. For example, Vav and
p115rhoGEF both contain DH and/or PH
domains, but they also contain distinct
regulatory and localization elements.
Have GEF domains diverged from a
common ancestor?
Even though all regulatory G proteins
appear to belong to a single, ancient
family of proteins, GEF domains do not;
the different classes of GEFs appear to
have arisen independently. The structures
of GEFs belonging to the sec7, DH, cdc25,
RCC1 (acts on Ran) and EfTu families
have been determined, and each has been
shown to adopt a different threedimensional fold (Fig. 1). The GPCR GEFs
constitute a distinct family of integral
membrane proteins.
How do they work?
Although the specific molecular
mechanisms differ, GEFs use a common
approach to release nucleotide. G proteins
share characteristic and well-conserved
catalytic sites, composed of a P-loop that
enfolds the nucleotide phosphates, together
with two mobile switch segments involved
in catalysis and effector recognition. The
very strong binding of guanine di- and
trinucleotides to G proteins arises, in part,
(b) Ras-cdc25 domain of Human SOS1 (1BKD); (c) DH–PH domain from Human SOS1
(1DBH), (d) Human Ran-GEF RCC1 (1A12); (e) Bovine rhodopsin (1F88).
http://tibs.trends.com 0968-0004/01/$ – see front matter © 2001 Elsevier Science Ltd. All rights reserved. PII: S0968-0004(01)01818-7
Forum
from a network of interactions between the
nucleotide phosphates and the P-loop, and
between a magnesium cation and one of
the switches. The four structures of GEF–
G-protein complexes that have been
determined are consistent with a scheme in
which the GEF disrupts the magnesium ion
binding site in the G protein, with a
concomitant rearrangement of one or both
switch segments4,5. Some GEFS also
disrupt the γ-phosphate binding site and the
P-loop (Fig. 2). By contrast, the guanine
binding site is maintained, allowing the
formation of a metastable ternary complex.
GDP diffuses from the active site, possibly
following an isomerization step6, leaving an
‘open’binary complex that is capable of
accepting nucleotide. Because the
intracellular concentration of GTP is
approximately tenfold greater than that of
GDP, an active, GTP-bound G protein is
the usual product of an encounter between a
G protein and its GEF. It is interesting to
note that, for ARF1, interaction with the
membrane is essential for the sec7catalyzed nucleotide exchange mechanism7.
How are GEFs regulated?
GEFs are the recipients of the regulatory
signals that trigger G-protein-coupled
pathways. Regulation of GEFs might
involve their specific localization, as well as
direct activation by protein–protein
interactions. Information gleaned from
studies of Ras-directed GEFs is instructive
for our understanding of these
mechanisms8. There are several distinct
families of Ras-GEF and, although all
members of these families contain cdc25
domains, each is responsive to different sets
of cellular inputs. SOS, for example,
contains an SH3 domain, facilitating the
localization of this GEF to the plasma
membrane (where it activates Ras) upon
receptor tyrosine kinase stimulation.
Members of the CalDEG-GEF family of Ras
GEFs possess EF-hand calcium binding
domains as well as diacylglycerol-binding
domains similar to those found in protein
kinase C. These GEFs are, therefore,
potentially responsive to phospholipase C
activation. Members of the Rap-directed
family of GEFs are regulated by cAMP
and are thus subject to regulation through
G-protein activation of adenylyl cyclase9.
Indeed, several examples of G-proteinregulated GEFs have emerged. For
example, both Ral-GDF, which contains a
cdc25 domain, and RIN1 (B. Horazdovsky,
pers. commun.), a Vps9 domain-containing
http://tibs.trends.com
TRENDS in Biochemical Sciences Vol.26 No.4 April 2001
267
Fig. 2. Ejection of GDP from Ras by SOS: left, the Ras•GDP•Mg2+ complex (4Q21), with GDP and Mg2+ in red, switch
regions in magenta and P-loop in blue; right, the Ras–SOS1 complex with the Ras-interacting loops of the cdc25
domain shown in green (1BKD).
GEF for Rab5, are effectors of Ras. Such
mechanisms allow a signal to diverge into
multiple pathways10. Signaling networks
also involve indirect connections. For
example, Rho-family GEFs Vav and Tiam
contain PH domains directly C-terminal to
their GEF domains. These phosphoinositolbinding domains might not only function as
membrane localization modules, but might
also potentiate the GEF activity of the
neighboring DH domain. In any case, PH
domain-containing GEFs are potentially
responsive to Ras-activated phosphoinositol
3-kinase. Novel members of the Rho familydirected GEFs have been identified that are
regulated by heterotrimeric G proteins. The
best characterized of these, p115rhoGEF, is
activated by G13α upon stimulation of
lysophosphatidic acid receptors11.
Remarkably, p115rhoGEF contains an
RGS (regulator of G-protein signaling)
domain that stimulates the GTPase
activity of, and thereby inactivates, its
own stimulator, G13α.
Where are the big challenges in GEF
research?
Much remains to be understood about the
molecular mechanisms of GEF action. For
example, it is still a mystery how GPCRs
work. G-protein α subunits do not, in
general, require a magnesium ion as a
cofactor for GDP binding. Therefore, we
can expect fundamental differences in the
way GPCRs promote nucleotide exchange
compared with other GEFs. Nor is it
understood how or, in some cases, to what
extent, GEFs are stimulated by their own
domains (e.g. cAMP-binding and PH) or
other G proteins. Perhaps the most
intriguing and far-reaching challenge is to
understand the complex and highlycoupled network of signaling events in
which GEFs participate. The elaborate
suites of regulatory modules with which
many GEFs are adorned suggest that
G-protein activation is localized,
contingent and temporally defined.
References
1 Cerione, R.A. and Zheng, Y. (1996) The Dbl family
of oncogenes. Curr. Opin. Cell Biol. 8, 216–222
2 Boguski, M.S. and McCormick, F. (1993) Proteins
regulating Ras and its relatives. Nature
366, 643–654
3 Roth, M.G. (1999) Snapshots of ARF1:
implications for mechanisms of activation and
inactivation. Cell 97, 149–152
4 Sprang, S.R. and Coleman, D.E. (1998) Invasion of
the nucleotide snatchers: structural insights into
the mechanism of G protein GEFs. Cell 95, 155–158
5 Worthylake, D.K. et al. (2000) Crystal structure of
Rac1 in complex with the guanine nucleotide
exchange region of Tiam1. Nature 408, 682–688
6 Hutchinson, J.P. and Eccleston, J.F. (2000)
Mechanism of nucleotide release from Rho by the
GDP dissociation stimulator protein.
Biochemistry 39, 11348–11359
7 Cherfils, J. and Chardin, P. (1999) GEFs: structural
basis for their activation of small GTP-binding
proteins. Trends Biochem. Sci. 24, 306–311
8 Bar-Sagi, D. and Hall, A. (2000) Ras and Rho
GTPases: a family reunion. Cell 103, 227–238
9 de Rooij, J. et al. (1998) Epac is a Rap1 guaninenucleotide-exchange factor directly activated by
cyclic AMP. Nature 396, 474–477
10 Henry, D.O. et al. (2000) Ral GTPases contribute
to regulation of cyclin D1 through activation of
NF-κB. Mol. Cell. Biol. 20, 8084–8092
11 Hart, M.J. et al. (1998) Direct stimulation of the
guanine nucleotide exchange activity of p115
RhoGEF by Gα13. Science 280, 2112–2114
Stephen Sprang
Howard Hughes Medical Institute, Dept of
Biochemistry, University of Texas, Southwestern
Medical Center, 75390-9050, USA.
e-mail: [email protected]