scispace - formally typeset
Open AccessJournal ArticleDOI

Duration of nuclear NF-κB action regulated by reversible acetylation

Reads0
Chats0
TLDR
It is demonstrated that the RelA subunit of NF-κB is subject to inducible acetylation and that acetylated forms of RelA interact weakly, if at all, with IκBα.
Abstract
The nuclear expression and action of the nuclear factor kappa B (NF-κB) transcription factor requires signal-coupled phosphorylation and degradation of the IκB inhibitors, which normally bind and sequester this pleiotropically active factor in the cytoplasm. The subsequent molecular events that regulate the termination of nuclear NF-κB action remain poorly defined, although the activation of de novo IκBα gene expression by NF-κB likely plays a key role. Our studies now demonstrate that the RelA subunit of NF-κB is subject to inducible acetylation and that acetylated forms of RelA interact weakly, if at all, with IκBα. Acetylated RelA is subsequently deacetylated through a specific interaction with histone deacetylase 3 (HDAC3). This deacetylation reaction promotes effective binding to IκBα and leads in turn to IκBα-dependent nuclear export of the complex through a chromosomal region maintenance-1 (CRM-1)–dependent pathway. Deacetylation of RelA by HDAC3 thus acts as an intranuclear molecular switch that both controls the duration of the NF-κB transcriptional response and contributes to the replenishment of the depleted cytoplasmic pool of latent NF-κB–IκBα complexes.

read more

Content maybe subject to copyright    Report

restore LexA-VP16 destruction in Met30-null
cells (Fig. 3A), the fusion did rescue tran-
scriptional activation, restoring wild-type
levels of VP16 TAD activity (Fig. 3B, com-
pare VP16 and Ub-VP16 in the Met30-null
cells). Moreover, both Ub and the VP16 TAD
are required for transcriptional activation in
the Met30-null cells because Ub alone fused
to LexA (Ub-) did not activate transcription
(14 ). The observation that Met30’s role in
transcription can be complemented by fusion
of LexA-VP16 to Ub argues that Met30 co-
activates the VP16 TAD by signaling LexA-
VP16 ubiquitylation. Moreover, the metabol-
ic stability of the Ub-VP16 protein (Fig. 3A)
demonstrates that it is Met30-mediated ubiq-
uitylation, not destruction, that is required for
transcriptional activation.
The requirement of ubiquitylation for VP16
activator function reveals that the degron func-
tion of the VP16 TAD is intimately tied to its
ability to activate transcription. The link be-
tween these processes provides a simple expla-
nation for the frequent and intimate overlap of
TADs and degrons (3–5). This requirement for
ubiquitylation, which has not been observed in
vitro, reveals a function for Ub distinct from its
role in proteolysis (2). Recent evidence has
demonstrated that the 19S subunit of the protea-
some plays an essential role in transcriptional
elongation (15). Given the role of the 19S com-
plex as a Ub binding module (16 ), it is possible
that activator ubiquitylation serves to recruit the
19S complex to promoters, where the chaperone
functions of this complex promote transcription
elongation.
Although our data demonstrate that proteol-
ysis is not required for transcriptional activation,
it is important to note that Met30 does direct
LexA-VP16 destruction. This suggests that ac-
tivator destruction by the proteasome is a natural
consequence of ubiquitylation. Because of the
dual role of Ub in transcriptional activation and
activator destruction, therefore, we propose that
Ub “licenses” transcription factors by linking
their activity to their destruction. We imagine
that non-ubiquitylated activators are stable and
inactive. Interactions of an activator with a Ub-
ligase result in activator ubiquitylation, which
simultaneously activates the transcription factor
and primes it for destruction by the proteasome.
Given the large number of transcription factors
that are targeted for Ub-mediated proteolysis, it
is possible that many transcription factors are
regulated through this mechanism.
References and Notes
1. D. Thomas, M. Tyers, Curr. Biol. 10, R341 (2000).
2. A. Varshavsky, Trends Biochem. Sci. 22, 383 (1997).
3. S. E. Salghetti, S. Y. Kim, W. P. Tansey, EMBO J. 18,
717 (1999).
4. E. Molinari, M. Gilman, S. Natesan, EMBO J. 18, 6439
(1999).
5. S. E. Salghetti, M. Muratani, H. Wijnen, B. Futcher, W. P.
Tansey, Proc. Natl. Acad. Sci. U.S.A. 97, 3118 (2000).
6. S. J. Triezenberg, R. C. Kingsbury, S. L. McKnight,
Genes Dev. 2, 718 (1988).
7. R. Brent, M. Ptashne, Cell 43, 729 (1985).
8. D. Skowyra, K. L. Craig, M. Tyers, S. J. Elledge, J. W.
Harper, Cell 91, 209 (1997).
9. Because the VP16 TAD contains no lysine residues,
LexA-VP16 ubiquitylation probably occurs within the
LexA protein.
10. A. Barberis, et al., Cell 81, 359 (1995).
11. Met30 (–) cells expressing LexA–VP16 grow at an
identical rate to Met30 () cells expressing this
protein. The indistinguishable growth rates of these
cells support the argument that LexA-VP16 does not
globally suppress (i.e., squelch) transcription in the
Met30-null background.
12. R. Li et al., Mol. Cell. Biol. 18, 1296 (1998).
13. Y. Marahrens, B. Stillman, Science 255, 817 (1992).
14.
The finding that the VP16 TAD is still required for
transcriptional activation in this setting demonstrates
that VP16 TAD function is not globally suppressed in the
absence of Met30. Together with the results of the
plasmid stability analysis (Fig. 2C), this finding demon-
strates that the VP16 TAD retains at least part of its
activity in the Met30-null cells.
15. A. Ferdous, F. Gonzalez, L. Sun, T. Kodadek, S. A.
Johnston, Mol. Cell 7, 981 (2001).
16. D. Voges, P. Zwickl, W. Baumeister, Annu. Rev. Bio-
chem. 68, 1015 (1999).
17. E. E. Patton et al., EMBO J. 19, 1613 (2000).
18. A. Rouillon, R. Barbey, E. E. Patton, M. Tyers, D.
Thomas, EMBO J. 19, 282 (2000).
19. E. S. Johnson, P. C. Ma, I. M. Ota, A. Varshavsky,
J. Biol. Chem. 270, 17442 (1995).
20.
Sequences encoding the VP16, Myc, and Cln3 degrons
were inserted into pRJR238 (10), modified to include an
in-frame hemagglutinin (HA)-epitope tag ( pRJR238HA).
The resulting LexA-fusion proteins were expressed in
CC849-1B (MAT a, his3, leu2, ura3, trp1, met4::TRP1) and
CC807-7D (MAT a, ade2, his3, leu2, ura3, trp1,
met4::TRP1, met30::LEU2) yeast (17), each carrying an
integrated LexA–-galactosidase reporter (10). Pulse-
chase analysis was performed as described (3).
21. LexA fusion proteins were expressed in CC849-1B,
along with either glutathione S-transferase (GST ) or
GST-Met30 (18) under the control of the GAL1 pro-
moter. Log-phase cultures were induced with galac-
tose, proteins were harvested, and GST proteins were
collected on glutathione agarose (18).
22. J. Spence et al., Cell 102, 67 (2000).
23. LexA-VP16 appears to be modified in Met30 ()
cells. This modification is not due to ubiquitylation
because that species of LexA-VP16 does not react
with antibodies to ubiquitin.
24. A. Hecht, M. Grunstein, Methods Enzymol. 304, 399
(1999).
25.
CC894-1B and CC807-7D, which carry the indicated
pRJR238HA plasmids, were transformed with pARS1-
LEXA, which carries a URA3 marker and a modified
LexA-ARS1 DNA replication origin. Plasmid stability was
measured as described (13).
26. Ub coding sequences were polymerase chain reaction
(PCR)–amplified from yeast genomic DNA with two
modifications: (i) the carboxy-terminal glycine resi-
due was changed to alanine to prevent removal of
the Ub moiety by isopeptidases (19), and (ii) the
T7-epitope tag sequence was added at the carboxy
terminus of Ub.
27. For reagents, we thank D. Finley, W. Herr, R. Li, M.
Ptashne, and D. Thomas. We thank S. Grewal, R. Li, A.
Matapurkar, and V. Valmeekam for technical help.
A.A.C. is a George A. and Marjorie H. Anderson Fellow
of the Watson School of Biological Sciences and a
Howard Hughes Medical Institute Predoctoral Fellow.
W.P.T. is a Kimmel Scholar. Supported by the Cold
Spring Harbor Laboratory Cancer Center Support
Grant CA45508 and by USPHS grant CA-13106 from
the National Cancer Institute.
30 April 2001; accepted 2 July 2001
Published online 19 July 2001;
10.1126/science.1062079
Include this information when citing this paper.
Duration of Nuclear NF-B
Action Regulated by Reversible
Acetylation
Lin-feng Chen,
1
Wolfgang Fischle,
1
Eric Verdin,
1,2
Warner C. Greene
1,2,3
*
The nuclear expression and action of the nuclear factor kappa B (NF-B) tran-
scription factor requires signal-coupled phosphorylation and degradation of the IB
inhibitors, which normally bind and sequester this pleiotropically active factor in
the cytoplasm. The subsequent molecular events that regulate the termination of
nuclear NF-B action remain poorly defined, although the activation of de novo
IB gene expression by NF-B likely plays a key role. Our studies now demonstrate
that the RelA subunit of NF-B is subject to inducible acetylation and that acety-
lated forms of RelA interact weakly, if at all, with IB. Acetylated RelA is sub-
sequently deacetylated through a specific interaction with histone deacetylase 3
(HDAC3). This deacetylation reaction promotes effective binding to IB and leads
in turn to IB-dependent nuclear export of the complex through a chromosomal
region maintenance-1 (CRM-1)– dependent pathway. Deacetylation of RelA by
HDAC3 thus acts as an intranuclear molecular switch that both controls the
duration of the NF-B transcriptional response and contributes to the replenish-
ment of the depleted cytoplasmic pool of latent NF-B–IB complexes.
NF-B corresponds to an inducible transcrip-
tion factor complex that plays a pivotal role
in regulating the inflammatory, immune, and
anti-apoptotic responses in mammals (1, 2).
The prototypical NF-␬〉 complex, which cor-
responds to a heterodimer of p50 and RelA
subunits, is sequestered in the cytoplasm by
its assembly with a family of inhibitory pro-
teins termed the IBs (1). Stimulus-induced
phosphorylation of two NH
2
-terminal serines
in the IBs, mediated by a macromolecular
IB kinase complex (IKK) (3), triggers the
R EPORTS
www.sciencemag.org SCIENCE VOL 293 31 AUGUST 2001 1653

rapid ubiquitination and subsequent degrada-
tion of this inhibitor by the 26S proteasome
complex. The liberated NF-␬〉 heterodimer
rapidly translocates into the nucleus, where it
engages cognate ␬〉 enhancer elements and
alters gene expression. The NF-B complex
also recruits the p300/CBP and P/CAF coac-
tivators, which participate in the activation of
target gene transcription (46 ). Phosphoryl-
ation of an NH
2
-terminal site in RelA by
protein kinase A facilitates NF-B assembly
with CBP/p300 (7 ). Both CBP and p300
contain histone acetyltransferase activity that
has been implicated in the regulation of gene
expression. These effects involve acetylation
of core histones leading to changes in chro-
matin structure (813) as well as direct acet-
ylation of select host transcription factors like
Fig. 1. (A) TSA en-
hances TNF-–medi-
ated activation of ␬〉-
luciferase gene expres-
sion. 293T cells were
transfected with B-
luciferase reporter
plasmid DNA (0.1 g)
and luciferase activity
was measured (19) af-
ter treatment with
TSA and TNF- (10
ng/ml). Results repre-
sent cumulated data
from three indepen-
dent transfections. (B)
TSA enhances TNF-
induction of nuclear
NF-B DNA-binding
activity. NF-B DNA-
binding activity was
assessed in electro-
phoretic mobility shift
assays (EMSA) with a
32
P-radiolabeled con-
sensus B enhancer
oligonucleotide (5-
AGTTGAGGGGACT T-
TCCCAGGC-3) (up-
per panel). Supershift-
ing this complex with anti-RelA (SC-109, Santa Cruz Biotechnology,
Santa Cruz, CA) and anti-p50 (SC-1190, Santa Cruz) is shown in lanes
7 and 8. Comparability of the various nuclear extracts was assessed
by EMSA with a
32
P-radiolabeled Sp1 probe (Promega) (lower panel).
(C) RelA expression is sustained in the nuclei of TNF-stimulated
cells in the presence of TSA. HeLa cells were incubated in the presence
(lanes 7 to 12) or absence (lanes 1 to 6) of TSA (400 nM) for 1 hour
before stimulation with TNF- (20 ng/ml) for 30 min. The cells were
chased for the indicated time periods with medium alone (lanes 1
to 6) or medium containing TSA (400 nM) (lanes 7 to 12). Nuclear
extracts from each culture were immunoblotted with anti-RelA and
visualized with an enhanced chemiluminescence reagent (Amersham).
(D) HDAC3, but not HDAC1, 2, 4, 5, or 6, inhibits TNF- activation
of B-luciferase activity. Expression levels of each HDAC determined
by anti-Flag immunoblotting of the cell lysates are shown in the
insets.
Fig. 2. (A) RelA is acetylated in vivo. COS-7 cells were
cotransfected with expression vectors encoding T7-RelA
and HDAC1 or HDAC3. Acetylation levels of RelA deter-
mined by [
3
H]sodium acetate labeling (19) are shown in
the upper panel. The total amount of T7-RelA in the
individual samples is shown in the lower panel. (B) Endog-
enous RelA is inducibly acetylated after TNF- stimulation.
Acetylation of endogenous RelA was assessed by [
3
H]so-
dium acetate labeling (19) and is shown in the upper panel.
The level of RelA present in each of the two immunopre-
cipitates is shown in the lower panel. (C) RelA is inducibly
acetylated by p300 in vivo. 293T cells were cotransfected
with T7-RelA and IB in the presence or absence of
expression plasmids encoding p300. Cell cultures were
stimulated with TNF- for the indicated time periods, and
acetylation levels of RelA were analyzed by immunoblot-
ting with antibody to acetylated lysine (Cell Signaling,
Beverly, MA). The level of T7-RelA present in each of the
immunoprecipitates is also shown in the lower panel.
R EPORTS
31 AUGUST 2001 VOL 293 SCIENCE www.sciencemag.org1654

p53, GATA-1, and E2F, which alters their
biological function (9, 10).
Although much progress has been made in
understanding the biochemical events that
underlie NF-B induction, much less is
known about how activated nuclear forms of
NF-B are negatively regulated, ensuring a
transient transcriptional response. Prior stud-
ies have demonstrated that the IB gene is
induced by NF-B(1416 ) and that the de
novo expression of IB proteins, which dis-
play nucleocytoplasmic shuttling properties,
participates in a negative feedback system
limiting the NF-B transcriptional response
(17, 18). We now demonstrate that nuclear
RelA is subject to reversible acetylation and
that this posttranslational modification plays
a pivotal role in NF-B regulation by gov-
erning IB binding to RelA and the nuclear
export of the NF-B complex.
We first observed that trichostatin A
(TSA), a specific inhibitor of the multiple
histone deacetylases (HDACs) (11), en-
hanced tumor necrosis factor (TNF)-–in-
duced, but not basal, expression of a B-
luciferase reporter gene (Fig. 1A). In addi-
tion, TSA enhanced nuclear NF-B DNA
binding after TNF- stimulation but did not
itself stimulate NF-B binding (Fig. 1B).
TSA also did not alter the binding of the
constitutively expressed Sp1 transcription
factor in either the presence or absence of
TNF- (Fig. 1B, lower panel). Immunoblot-
ting of the nuclear extracts with antibodies to
RelA (anti-RelA) revealed sustained nuclear
RelA expression in the presence of TSA (Fig.
1C). Together, these results indicate that TSA
enhances TNF-–induced NF-B DNA bind-
ing, likely by prolonging intranuclear expres-
sion of RelA.
TSA broadly inhibits the action of the
HDACs. HDACs function not only to deacety-
late core histones leading to repressive changes
in chromatin structure but also to deacetylate
various host transcription factors, altering their
transcriptional activity (1013). We assessed
potential inhibitory effects of various HDACs
on NF-B action by cotransfecting 293T cells
with expression vectors encoding HDAC1, 2, 3,
4, 5, or 6 and a B-luciferase reporter followed
by stimulation with TNF- for 5 hours (Fig.
1D). Although each of the HDACs was com-
parably expressed, only HDAC3 inhibited
TNF-–induced B-luciferase activity (lanes 7
and 8). This inhibitory effect of HDAC3 was
abrogated in the presence of TSA, indicating
that the deacetylase function of HDAC3 was
required for these biological effects (Web fig.
1) (19). Consistent with this action of HDAC3,
we found that the RelA subunit of NF-Bis
acetylated in vivo (Fig. 2A, lane 2, upper pan-
el). RelA acetylation was abolished in the pres-
ence of HDAC3 (lane 5) but not HDAC1 (lane
3). The addition of TSA to the HDAC3-ex-
pressing cultures was associated with the detec-
tion of acetylated RelA (lane 4). When HeLa
cells were incubated in [
3
H]sodium acetate and
stimulated with TNF- or medium for 30 min,
signal-induced acetylation of endogenous RelA
was similarly detected (Fig. 2B). Together,
these studies reveal that NF-B is subject to
reversible acetylation and that HDAC3 plays a
central role in its deacetylation.
The potential participation of the p300,
1
Gladstone Institute of Virology and Immunology,
2
Department of Medicine,
3
Department of Microbiol-
ogy and Immunology, University of California, San
Francisco, CA 94141, USA.
*To whom correspondence should be addressed. E-
mail: wgreene@gladstone.ucsf.edu
Fig. 3. (A) HDAC3 and RelA physically assemble in vivo. COS-7 cells were
transfected with the indicated combinations of cDNAs encoding Flag epi-
tope–tagged HDAC1 or HDAC3 and T7-RelA, and anti-T7 immunoprecipi-
tations were performed followed by anti-Flag immunoblotting (19). Levels of
expression of HDAC1-Flag, HDAC3-Flag, and T7-RelA are shown in the lower
two panels. (B) The physical association of endogenous RelA and endoge-
nous HDAC3 in HeLa cells. Whole-cell lysates from HeLa cells were immu-
noprecipitated with polyclonal anti-HDAC1 or anti-HDAC3 followed by
immunoblotting with monoclonal anti-RelA (F-6, Santa Cruz). (C) HDAC3
inhibits TNF-–induced nuclear expression of RelA. HeLa cells were trans-
fected with control (lanes 1 to 4) or HDAC3-expression vector DNA and
cultured for 24 hours, followed by stimulation with TNF- (10 ng/ml) for 0, 10, 30, or 90 min. Nuclear extracts were prepared and EMSAs performed
as described in Fig. 1B (panels 1 and 2). TNF-–induced degradation of I␬〉␣ was analyzed in the cytoplasmic extracts of these cultures by
immunoblotting with anti-IB (C-21, Santa Cruz) (panel 3). Levels of nuclear RelA were assessed by immunoblotting nuclear extracts with specific
anti-RelA (panel 4). The faster migrating bands reactive with the anti-RelA likely correspond to RelA degradation products and show similar changes
in levels of nuclear expression. (D) HDAC3 stimulates nuclear export of RelA via a leptomycin B–sensitive pathway. GFP-RelA principally localized in
the nucleus when expression plasmid DNA was transfected into HeLa cells. Coexpression of HDAC3 and GFP-RelA resulted in a cytoplasmic expression
pattern of GFP-RelA; HDAC3 and GFP-RelA plasmid DNA was transfected at a 6:1 ratio to ensure that all GFP-RelA–expressing cells also contained
HDAC3. Coexpression of HDAC1 and GFP-RelA did not alter the nuclear pattern of GFP-RelA epifluorescence. Treatment of the GFP-RelA and
HDAC3–expressing cell cultures with TSA (800 nM, 5 hours) resulted in a nuclear pattern of expression of GFP-RelA. Treatment of GFP-RelA
and HDAC3– expressing cultures with leptomycin B (LMB; 20 nM, 2 hours) produced a nuclear pattern of GFP-RelA epifluorescence. Average
percentages of cells displaying the depicted phenotype are shown in the lower right corner of each panel. Results are derived from inspection
of at least 200 transfected cells present in multiple microscopic fields from two independent experiments.
R EPORTS
www.sciencemag.org SCIENCE VOL 293 31 AUGUST 2001 1655

CBP, and P/CAF acetyltransferases in the
acetylation of RelA was next investigated.
When 293T cells were cotransfected with
RelA, IB, and p300, and stimulated with
TNF-, inducible acetylation of RelA was
detected (Fig. 2C). Coexpression of either
p300 or CBP, but not P/CAF, with RelA
produced a dose-dependent acetylation of T7-
RelA [Web fig. 2 (19)]. Together, these find-
ings demonstrate that endogenous RelA is
acetylated in a signal-coupled manner likely
mediated by p300 and CBP.
Coimmunoprecipitation of T7-RelA and
HDAC3 in COS-7 cells suggests a physical
interaction between these two proteins in
vivo (Fig. 3A). In contrast, no interaction
between RelA and HDAC1 was detected de-
spite comparable levels of both HDACs be-
ing expressed (Fig. 3A). Moreover, anti-
HDAC3 effectively coimmunoprecipitated
endogenous RelA from HeLa cells, whereas
only trace amounts of RelA immunoprecipi-
tated with anti-HDAC1 (Fig. 3B). By using
coimmunoprecipitation assays and the mam-
malian two-hybrid system (20), we found that
NH
2
-terminal regions of both HDAC3 and
RelA are required for the assembly of these
two proteins in vivo [Web fig. 3) (19)].
Studies were next performed to define the
molecular basis for HDAC3-mediated inhibi-
tion of TNF- activation of NF-B. Expression
of HDAC3 in HeLa cells (50% transfection
efficiency) diminished both NF-B DNA-bind-
ing activity (Fig. 3C, panel 1) and levels of
nuclear RelA ( panel 4). In contrast, HDAC3
expression did not alter TNF-–induced degra-
dation of IB occurring in the cytoplasm
( panel 3), nor did it markedly change the levels
of Sp1 DNA-binding activity ( panel 2). To-
gether with the results presented in Fig. 1C,
these findings raise the possibility that HDAC3
may regulate the intracellular trafficking of
RelA. A precedent for acetylation influencing
the intracellular trafficking of a transcription
factor is provided by recent studies on hepato-
cyte nuclear factor-4 and class II transactivator
(CIITA) (21, 22). To monitor potential effects
of HDAC3 on the intracellular trafficking of
RelA, we expressed green fluorescent protein–
RelA fusion proteins (GFP-RelA) in HeLa cells
in the presence and absence of HDAC3 (Fig.
3D). When expressed alone or with HDAC1,
GFP-RelA localized to the nucleus, whereas the
coexpression of HDAC3 resulted in a predom-
inantly cytoplasmic pattern of GFP-RelA ex-
pression. This cytoplasmic relocalization of
GFP-RelA induced by HDAC3 did not occur in
the presence of TSA. These effects of HDAC3
appeared to result from nuclear export of GFP-
RelA because the addition of leptomycin B, a
known inhibitor of CRM-1/exportin-1– depen-
dent nuclear export (23), preserved the nuclear
pattern of GFP-RelA epifluorescence. These
findings suggest that HDAC3-mediated
deacetylation of RelA promotes its export from
the nucleus to the cytoplasm.
Among the target genes activated by NF-B
is the IB gene (1416 ). The resultant de
novo synthesis of IB serves to replenish the
intracellular stores of this inhibitor depleted
during the course of NF-B activation. IB
also displays nucleocytoplasmic shuttling prop-
erties and likely retrieves nuclear NF-B com-
plexes, thereby contributing to the termination
of the NF-B transcriptional response (17, 18).
We next examined whether the acetylation sta-
tus of RelA regulates its assembly with IB.
The acetylation of RelA induced by the coex-
pression of increasing amounts of p300 was
associated with markedly diminished binding
of RelA to GST-IB matrices (Fig. 4A, lanes
3 and 4). However, the coexpression of increas-
ing amounts of HDAC3 in the presence of p300
restored RelA binding to GST-IB (Fig. 4A,
lanes 5 and 6). Immunoblotting of the T7-RelA
proteins with antibodies to acetylated lysine
confirmed dose-related increases in RelA acet-
ylation by p300 in lanes 3 and 4 and dose-
related deacetylation of RelA by HDAC3 in
lanes 5 and 6 (24 ). These findings suggest that
acetylation of RelA prevents IB binding,
whereas deacetylation of RelA by HDAC3
stimulates IB binding.
To test whether the HDAC3-induced nu-
clear export of RelA is dependent on IB,
we studied the subcellular localization of
GFP-RelA in murine embryo fibroblasts
(MEFs) isolated from wild-type or IB
–/–
mice produced by targeted gene disruption
Fig. 4. (A) Deacetylated RelA
displays greater IB-binding
activity than acetylated RelA.
293T cells were cotransfected
with T7-RelA and p300 in the
presence or absence of
HDAC3 expression vector
DNA as indicated. Twenty-
four hours later, 50 l of each
whole-cell lysate was incubat-
ed with GST-IB (1.0 g).
The levels of RelA captured by
the GST-IB matrix under
each condition were assessed
by immunoblotting with anti-
T7 (upper panel). The total
amounts of GST-IB and
RelA present in the reaction
mixtures are shown in the
lower two panels. (B) HDAC3
does not stimulate GFP-RelA
nuclear export in IB-defi-
cient MEFs. MEF cells from
wild-type and IB
–/–
mice
were transfected with GFP-
RelA (0.3 g) ( panels a and c)
or GFP-RelA (0.3 g) and
HDAC3 (1.7 g) ( panels b
and d) expression vector DNA. The IB
/
MEFs were also recon-
stituted with limiting amounts of IB expression vector (0.1 g) in
the absence and presence of HDAC3 ( panels e and f ). Average
percentages of cells displaying the depicted phenotype derived from
inspection of at least 120 cells present in multiple microscopic fields
from two independent experiments are shown in the lower right
corner of each panel. (C) Schematic model for the role of HDAC3-
mediated deacetylation of RelA as an intranuclear molecular switch
promoting IB binding and IB-dependent nuclear export of the
NF-B complex. This deacetylation-controlled response both leads to
the termination of the NF-B transcriptional response and aids in
reestablishing latent cytoplasmic forms of NF-B bound to IB,
thereby preparing the cell to respond to the next NF-B–inducing
stimulus.
R EPORTS
31 AUGUST 2001 VOL 293 SCIENCE www.sciencemag.org1656

(25) (Fig. 4B). In wild-type MEFs, GFP-
RelA was principally expressed in the nucle-
us ( panel a), whereas the coexpression of
HDAC3 induced a cytoplasmic pattern of
GFP-RelA epifluorescence ( panel b). How-
ever, a very different pattern was obtained in
the IB
–/–
MEFs. Whereas GFP-RelA also
exhibited a nuclear pattern of epifluores-
cence, the coexpression of HDAC3 in these
IB
–/–
MEFs failed to induce a cytoplasmic
redistribution of RelA ( panel d). Reconstitu-
tion of these IB
–/–
cells by transfection
with small quantities of an IB expression
vector restored HDAC3-induced cytoplasmic
expression of the GFP-RelA protein (panel
f ). In the absence of HDAC3, GFP-RelA
remained principally nuclear, indicating that
the levels of IB expressed were not suffi-
cient on their own to produce cytoplasmic
sequestration of GFP-RelA in these IB
–/–
MEFs ( panel e). These results indicate that
IB is required for the nuclear export of
deacetylated forms of RelA, which display
increased binding of IB.
These findings reveal a new mechanism
through which nuclear NF-B function is regu-
lated (Fig. 4C). RelA is subject to stimulus-
coupled acetylation likely mediated through the
p300 and CBP coactivators. One biological con-
sequence of this modification is that acetylated
RelA becomes a very poor substrate for binding
by newly synthesized IB. Whether p50 or
perhaps IB are similarly subject to biologi-
cally important acetylation/deacetylation reac-
tions remains to be explored. Our studies iden-
tify acetylated RelA as a novel nonhistone sub-
strate of HDAC3. As such, HDAC3-mediated
deacetylation functions as an intranuclear mo-
lecular switch that when activated initiates a
series of events culminating in the termination
of the NF-B transcriptional response. The
IB-dependent nuclear export of the HDAC3-
deacetylated RelA-containing complexes also
serves to replenish the depleted cytoplasmic
pool of latent NF-B–IB complexes needed
for the next inductive NF-B response in these
cells.
References and Notes
1. A. S. Baldwin Jr., Annu. Rev. Immunol. 14, 649 (1996).
2. S. Ghosh, M. J. May, E. B. Kopp, Annu. Rev. Immunol.
16, 225 (1998).
3. M. Karin, Oncogene 18, 6867 (1999).
4. N. D. Perkins et al., Science 275, 523 (1997).
5. M. O. Hottiger, L. K. Felzien, G. J. Nabel, EMBO J. 17,
3124 (1998).
6. K. A. Sheppard et al., Mol. Cell. Biol. 19, 6367 (1999).
7. H. Zhong, R. E. Voll, S. Ghosh, Mol. Cell 1, 661 (1998).
8. A. Imhof, A. P. Wolffe, Curr. Biol. 8, 422 (1998).
9. D. E. Sterner, S. L. Berger, Microbiol. Mol. Biol. Rev.
64, 435 (2000).
10. M. T. Kouzarides, EMBO J. 19, 1176 (2000).
11. H. Kuo, C. D. Allis, Bioessays 20, 615 (1998).
12. W. D. Cress, E. Seto, J. Cell Physiol. 184, 1 (2000).
13. H. H. Ng, A. Bird, Trends Biochem. Sci. 25, 121 (2000).
14. S. C. Sun, P. A. Ganchi, D. W. Ballard, W. C. Greene,
Science 259, 1912 (1993).
15. K. Brown, S. Park, T. Kanno, G. Franzoso, U. Siebenlist,
Proc. Natl. Acad. Sci. U.S.A. 90, 2532 (1993).
16. A. A. Beg, T. S. Finco, P. V. Nantermet, A. S. Baldwin
Jr., Mol. Cell. Biol. 13, 3301 (1993).
17. F. Arenzana-Seisdedos et al., Mol. Cell. Biol. 15, 2689
(1995).
18. F. Arenzana-Seisdedos et al., J. Cell Sci. 110, 369
(1997).
19. Supplementary figures and information on experi-
mental protocols are available on Science Online at
www.sciencemag.org/cgi/content/full/293/5535/
1653/DC1.
20. H. Bogerd, W. C. Greene, J. Virol. 67, 2496 (1993).
21. E. Soutoglou, N. Katrakili, I. Talianidis, Mol. Cell 5,
745 (2000).
22. C. Spilianakis, J. Papamatheakis, A. Kretsovali, Mol.
Cell. Biol. 20, 8489 (2000)
23. M. Yoshida, S. Horinouchi, Ann. N. Y. Acad. Sci. 886,
23 (1999).
24. L.-F. Chen, W. C. Greene, unpublished data.
25. J. F. Klement et al., Mol. Cell. Biol. 16, 2341 (1996).
26. We thank R. Goodman (Vollum Institute) and Y.
Nakatani (NIH) for reagents; J. F. Klement ( Jefferson
Medical College) for providing the IB
/
MEF cells;
and A. O’Mahony for helping establish cultures of
these cells. We also thank J. Carroll for assistance in
the preparation of the figures and R. Givens for
assistance in the preparation of the manuscript. Sup-
ported in part by funds provided by the J. David
Gladstone Institutes, Pfizer, and the University of
California, San FranciscoGladstone Institute of Vi-
rology and Immunology Center for AIDS Research
(P30MH59037).
8 May 2001; accepted 3 July 2001
Allosteric Activation of a
Spring-Loaded Natriuretic
Peptide Receptor Dimer by
Hormone
Xiao-lin He, Dar-chone Chow, Monika M. Martick,
K. Christopher Garcia*
Natriuretic peptides (NPs) are vasoactive cyclic-peptide hormones important in
blood pressure regulation through interaction with natriuretic cell-surface re-
ceptors. We report the hormone-binding thermodynamics and crystal struc-
tures at 2.9 and 2.0 angstroms, respectively, of the extracellular domain of the
unliganded human NP receptor (NPR-C) and its complex with CNP, a 22–amino
acid NP. A single CNP molecule is bound in the interface of an NPR-C dimer,
resulting in asymmetric interactions between the hormone and the symmet-
rically related receptors. Hormone binding induces a 20 angstrom closure
between the membrane-proximal domains of the dimer. In each monomer, the
opening of an interdomain cleft, which is tethered together by a linker peptide
acting as a molecular spring, is likely a conserved allosteric trigger for intra-
cellular signaling by the natriuretic receptor family.
The natriuretic peptides (NPs) are three ho-
mologous peptide hormones that play impor-
tant roles in the maintenance of cardiovascu-
lar homeostasis, blood pressure, and body
fluid regulation (i.e., natriuresis) (1). Collec-
tively, these hormones function as an endog-
enous counterbalance to the renin-angioten-
sin/aldosterone system, as well as the hypo-
thalamic/pituitary/adrenal axis. The three
members of this family are atrial (ANP) and
brain (BNP) natriuretic peptides, which are
produced by the heart; and CNP, which is
expressed in endothelial cells (1). ANP and
BNP are thought to be the primary regulators
of peripheral natriuretic activity; CNP is
present mainly in the brain. ANP, BNP, and
CNP are highly homologous (70% identi-
cal) and share as a common motif a 17–amino
acid loop formed by a disulfide bond (Fig. 1).
The lack of defined structure(s) in solution
and the questionable relevance of “lowest
energy” solution conformations to the recep-
tor-bound conformation of peptide hormones
mean that assessing the bioactive conforma-
tions of these peptides remains a general
problem (2).
The actions of the NPs are mediated by
three homologous single-transmembrane,
glycosylated cell-surface receptors (NPR-A,
-B, and -C) (35). These receptors possess
about 30% homologous extracellular ligand-
binding domains (ECDs) (450 amino acids)
with conserved topologies but possess differ-
ent downstream activation mechanisms.
NPR-C is the most promiscuous of the recep-
tors, binding to all NPs with high affinity,
whereas NPR-A and NPR-B are more specif-
ic for ANP and CNP, respectively (6 ). In the
cases of NPR-A and NPR-B, hormone bind-
ing to the ECDs results in the production of
intracellular cyclic guanosine 3,5-mono-
phosphate by a guanylyl-cyclase activity that
resides in the intracellular domains (5, 7 ). For
NPR-C, which represents over 95% of NPR
in vivo, ligand binding results in both inter-
nalization and degradation (i.e., clearance), as
well as signaling by heterotrimeric GTP-
R EPORTS
www.sciencemag.org SCIENCE VOL 293 31 AUGUST 2001 1657
Figures
Citations
More filters
Journal ArticleDOI

Signaling to NF-kappaB.

TL;DR: An overview of established NF-kappaB signaling pathways is provided with focus on the current state of research into the mechanisms that regulate IKK activation and NF- kappaB transcriptional activity.
Journal ArticleDOI

NF-kappaB regulation in the immune system.

TL;DR: The role of NF-κB proteins as potential therapeutic targets in clinical applications and their role in the immune system and inflammatory diseases are discussed.
Journal ArticleDOI

Missing Pieces in the NF-κB Puzzle

TL;DR: In this paper, a review of recent progress as well as unanswered questions regarding the regulation and function of NF-kappaB and IKK is presented, focusing on recent progress and unanswered questions.
Journal ArticleDOI

Anticancer activities of histone deacetylase inhibitors.

TL;DR: Recent advances in the understanding of the molecular events that underlie the anticancer effects of HDAC inhibitors are summarized and how such information could be used in optimizing the development and application of these agents in the clinic, either as monotherapies or in combination with other anticancer drugs are discussed.
Journal ArticleDOI

Modulation of NF-κB-dependent transcription and cell survival by the SIRT1 deacetylase

TL;DR: It is demonstrated that SIRT1, a nicotinamide adenosine dinucleotide‐dependent histone deacetylase, regulates the transcriptional activity of NF‐κB and activity augments apoptosis in response to TNFα.
References
More filters
Journal ArticleDOI

THE NF-κB AND IκB PROTEINS: New Discoveries and Insights

TL;DR: The transcription factor NF-κB has attracted widespread attention among researchers in many fields based on its unusual and rapid regulation, the wide range of genes that it controls, its central role in immunological processes, the complexity of its subunits, and its apparent involvement in several diseases.
Journal ArticleDOI

NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses

TL;DR: Recently, significant advances have been made in elucidating the details of the pathways through which signals are transmitted to the NF-kappa B:I kappa B complex in the cytosol and their implications for the study of NF-Kappa B.
Journal ArticleDOI

Acetylation of Histones and Transcription-Related Factors

TL;DR: This work detail these known factor acetyltransferase (FAT) substrates and the demonstrated or potential roles of their acetylation in transcriptional processes.
Journal ArticleDOI

Roles of histone acetyltransferases and deacetylases in gene regulation

TL;DR: Recent developments in histone acetylation and transcriptional regulation are reviewed, suggesting the possibility that internal lysine acetylations of multiple proteins exists as a rapid and reversible regulatory mechanism much like protein phosphorylation.
Journal ArticleDOI

Phosphorylation of NF-κB p65 by PKA Stimulates Transcriptional Activity by Promoting a Novel Bivalent Interaction with the Coactivator CBP/p300

TL;DR: Phosphorylation by PKA both weakens the interaction between the N- and C-terminal regions of p65 and creates an additional site for interaction with CBP/p300, which regulates the transcriptional activity of NF-kappa B by modulating its interaction withCBP/ p300.
Related Papers (5)
Frequently Asked Questions (1)
Q1. What are the contributions in this paper?

The link between these processes provides a simple explanation for the frequent and intimate overlap of TADs and degrons ( 3–5 ). Because of the dual role of Ub in transcriptional activation and activator destruction, therefore, the authors propose that Ub “ licenses ” transcription factors by linking their activity to their destruction. This suggests that activator destruction by the proteasome is a natural consequence of ubiquitylation.