ArticlePDF Available

The Glucocorticoid Receptor and Foxo1 Synergistically Activate the Skeletal Muscle Atrophy Associated Murf1 Gene

Authors:
  • Robert Bosch Center for Tumor Diseases

Abstract

The muscle specific ubiquitin E3 ligase MuRF1 has been implicated as a key regulator of muscle atrophy under a variety of conditions, such as during synthetic glucocorticoid treatment. FOXO class transcription factors have been proposed as important regulators of MuRF1 expression, but its regulation by glucocorticoids is not well understood. The MuRF1 promoter contains a near-perfect palindromic glucocorticoid response element (GRE) 200 base pairs upstream of the transcription start site. The GRE is highly conserved in the mouse, rat, and human genes along with a directly adjacent FOXO binding element (FBE). Transient transfection assays in HepG2 cells and C(2)C(12) myotubes demonstrate that the MuRF1 promoter is responsive to both the dexamethasone (DEX)-activated glucocorticoid receptor (GR) and FOXO1, whereas coexpression of GR and FOXO1 leads to a dramatic synergistic increase in reporter gene activity. Mutation of either the GRE or the FBE significantly impairs activation of the MuRF1 promoter. Consistent with these findings, DEX-induced upregulation of MuRF1 is significantly attenuated in mice expressing a homodimerization-deficient GR despite no effect on the degree of muscle loss in these mice vs. their wild-type counterparts. Finally, chromatin immunoprecipitation analysis reveals that both GR and FOXO1 bind to the endogenous MuRF1 promoter in C(2)C(12) myotubes, and IGF-I inhibition of DEX-induced MuRF1 expression correlates with the loss of FOXO1 binding. These findings present new insights into the role of the GR and FOXO family of transcription factors in the transcriptional regulation of the MuRF1 gene, a direct target of the GR in skeletal muscle.
doi:10.1152/ajpendo.00646.2007
295:785-797, 2008. First published Jul 8, 2008;Am J Physiol Endocrinol Metab
Holger M. Reichardt, J. David Furlow and Sue C. Bodine
David S. Waddell, Leslie M. Baehr, Jens van den Brandt, Steven A. Johnsen,
You might find this additional information useful...
60 articles, 26 of which you can access free at: This article cites http://ajpendo.physiology.org/cgi/content/full/295/4/E785#BIBL
including high-resolution figures, can be found at: Updated information and services http://ajpendo.physiology.org/cgi/content/full/295/4/E785
can be found at: AJP - Endocrinology and Metabolismabout Additional material and information http://www.the-aps.org/publications/ajpendo
This information is current as of October 22, 2008 .
http://www.the-aps.org/.
20814-3991. Copyright © 2005 by the American Physiological Society. ISSN: 0193-1849, ESSN: 1522-1555. Visit our website at
organization. It is published 12 times a year (monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD
publishes results of original studies about endocrine and metabolic systems on any level ofAJP - Endocrinology and Metabolism
on October 22, 2008 ajpendo.physiology.orgDownloaded from
The glucocorticoid receptor and FOXO1 synergistically activate the skeletal
muscle atrophy-associated MuRF1 gene
David S. Waddell,
1
Leslie M. Baehr,
1
Jens van den Brandt,
4
Steven A. Johnsen,
2,3
Holger M. Reichardt,
4
J. David Furlow,
1
* and Sue C. Bodine
1
*
1
Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California;
2
European Molecular
Biology Laboratory, Heidelberg;
3
Department of Molecular Oncology, Go¨ttingen Center for Molecular Biosciences;
and
4
Department of Cellular and Molecular Immunology, University of Go¨ttingen, Go¨ ttingen, Germany
Submitted 4 October 2007; accepted in final form 27 June 2008
Waddell DS, Baehr LM, van den Brandt J, Johnsen SA,
Reichardt HM, Furlow JD, Bodine SC. The glucocorticoid receptor
and FOXO1 synergistically activate the skeletal muscle atrophy-associ-
ated MuRF1 gene. Am J Physiol Endocrinol Metab 295: E785–E797,
2008. First published July 8, 2008; doi:10.1152/ajpendo.00646.2007.—
The muscle specific ubiquitin E3 ligase MuRF1 has been implicated as a
key regulator of muscle atrophy under a variety of conditions, such as
during synthetic glucocorticoid treatment. FOXO class transcription fac-
tors have been proposed as important regulators of MuRF1 expression,
but its regulation by glucocorticoids is not well understood. The MuRF1
promoter contains a near-perfect palindromic glucocorticoid response
element (GRE) 200 base pairs upstream of the transcription start site. The
GRE is highly conserved in the mouse, rat, and human genes along with
a directly adjacent FOXO binding element (FBE). Transient transfection
assays in HepG2 cells and C
2
C
12
myotubes demonstrate that the MuRF1
promoter is responsive to both the dexamethasone (DEX)-activated
glucocorticoid receptor (GR) and FOXO1, whereas coexpression of GR
and FOXO1 leads to a dramatic synergistic increase in reporter gene
activity. Mutation of either the GRE or the FBE significantly impairs
activation of the MuRF1 promoter. Consistent with these findings, DEX-
induced upregulation of MuRF1 is significantly attenuated in mice
expressing a homodimerization-deficient GR despite no effect on the
degree of muscle loss in these mice vs. their wild-type counterparts.
Finally, chromatin immunoprecipitation analysis reveals that both GR
and FOXO1 bind to the endogenous MuRF1 promoter in C
2
C
12
myo-
tubes, and IGF-I inhibition of DEX-induced MuRF1 expression corre-
lates with the loss of FOXO1 binding. These findings present new
insights into the role of the GR and FOXO family of transcription factors
in the transcriptional regulation of the MuRF1 gene, a direct target of the
GR in skeletal muscle.
forkhead transcription factor class O; muscle RING finger 1; gluco-
corticoid receptor
SKELETAL MUSCLE IS A DYNAMIC TISSUE that has the capacity to
continuously regulate its size in response to a variety of
external cues, including mechanical load, neural activity, hor-
mones/growth factors, stress, and nutritional status. In addition,
skeletal muscle serves as the most significant repository for
protein in the body, a source that is tapped to provide a pool of
amino acids for tissue repair and gluconeogenesis under con-
ditions of starvation and other metabolic stresses. Muscle loss
or “atrophy” occurs as the result of a number of disparate
conditions, including aging, immobilization, metabolic dis-
eases, cancer, and neurodegenerative diseases, and as a serious
side effect of therapeutic corticosteroid hormone treatment (15,
27, 32). The recently identified E3 ubiquitin ligase, muscle
RING finger 1 (MuRF1), is proposed to be a key regulator of
the atrophy process given that 1) it is expressed predominantly
in skeletal muscle (3), 2) it is upregulated under a variety of
atrophy conditions (3, 12, 25), and 3) deletion of the gene in
mice results in significant muscle sparing following denerva-
tion (3). Although the full physiological functions of MuRF1
are not yet known, it is often assumed that it functions in some
manner to regulate protein degradation since it is expressed
early in the atrophy process and its peak expression usually
occurs during maximum muscle loss. For example, MuRF1 has
been shown to play a direct role in myosin heavy chain
ubiquitination and degradation during synthetic glucocorticoid
treatment (5). However, MuRF1 may have additional impor-
tant functions in skeletal muscle, such as inhibition of protein
synthesis during starvation conditions (22) as well as regulat-
ing carbohydrate metabolism (16). Despite considerable inter-
est in MuRF1 as a regulator of skeletal muscle mass and
metabolism, there are limited data on the transcriptional regu-
lation of the MuRF1 gene.
Both natural and synthetic glucocorticoid hormones are
potent inducers of skeletal muscle atrophy (14). Glucocorti-
coids exert their physiological actions primarily via a nuclear
pathway to directly affect target gene transcription. Natural
glucocorticoids such as cortisol and corticosterone, as well as
synthetic glucocorticoids such as dexamethasone (DEX) and
prednisolone, exert their biological effects predominantly via
the glucocorticoid receptor (GR) (56). The GR is a member of
the nuclear receptor superfamily and acts as a ligand-dependent
transcription factor. Skeletal muscle expresses significant lev-
els of GR, as do cultured myotubes. In the absence of ligand,
the GR is found largely in the cytoplasm in a large complex
that includes chaperones such as heat shock protein 90. Upon
ligand binding, the GR becomes localized in the cell nucleus
and binds to DNA sequences called glucocorticoid response
elements (GREs). The consensus GRE sequence is AGAA-
CANNNTGTTCT, where two GRs bind to each six-nucleotide
half-site of the palindome as a homodimeric complex (2). The
three-base pair (bp) spacer sequence (NNN) can consist of any
nucleotide combination, although that particular sequence
length is critical. However, the perfect consensus sequence is
rarely found in native glucocorticoid-responsive promoters,
with two or more nucleotide differences often found in either
* J. D. Furlow and S. C. Bodine contributed equally to this study.
Address for reprint requests and other correspondence: J. D. Furlow or S. C.
Bodine, Section of Neurobiology, Physiology & Behavior, Univ. of California
Davis, One Shields Ave., Davis, CA 95616 (e-mail: jdfurlow@ucdavis.edu or
scbodine@ucdavis.edu).
The costs of publication of this article were defrayed in part by the payment
of page charges. The article must therefore be hereby marked advertisement
in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Am J Physiol Endocrinol Metab 295: E785–E797, 2008.
First published July 8, 2008; doi:10.1152/ajpendo.00646.2007.
0193-1849/08 $8.00 Copyright ©2008 the American Physiological Societyhttp://www.ajpendo.org E785
on October 22, 2008 ajpendo.physiology.orgDownloaded from
or both half-sites (45). In addition, GR may positively or
negatively influence transcription of target genes in a GRE-
and homodimer-independent manner via interaction with pro-
moter-bound STAT5, AP-1, or NF-B transcription factors
(reviewed in Ref. 50). Development of a mouse strain express-
ing a dimerization-deficient GR [GR homodimerization mutu-
ant (GR
dim
)] has allowed the identification of physiological
processes and specific gene expression that are dependent on
the classical GRE binding activity of the GR vs. an indirect
action through other transcription factors (38, 39).
Although the synthetic glucocorticoid DEX induces MuRF1
mRNA accumulation in vivo as well as in cultured myotubes
(3, 48), it is currently unknown whether the gene is directly
regulated by ligand-bound GR in vivo.DEX induction could
be mediated by the increased expression of other transcription
factors and associated coactivators that in turn bind to and
activate the MuRF1 promoter. For example, the class O type
forkhead transcription factors (FOXO), including FOXO1,
FOXO3a, and FOXO4, have recently been implicated as key
regulators of gene expression during skeletal muscle atrophy
(42, 48). FOXO1, FOXO3a, and FOXO4 are all expressed in
skeletal muscle (17), and FOXO1 and FOXO3a mRNA in
particular are upregulated during fasting and DEX treatment
(9, 25). Constitutively active FOXO proteins can activate the
endogenous MuRF1 gene (42, 48); however, there is no infor-
mation on the direct effect of FOXO proteins on the MuRF1
promoter.
Recently, it has been demonstrated that upregulation of
MuRF1 expression following DEX treatment or starvation of
C
2
C
12
cells can be suppressed by IGF-I (41, 42, 48). The
mechanism by which IGF-I is able to suppress MuRF1 tran-
scription is believed to be at least in part via the phosphatidyl-
inositol 3-kinase/Akt pathway. Akt phosphorylates members of
the FOXO class of forkhead transcription factors (40, 60), and
phosphorylated FOXOs are sequestered in the cytoplasm by
14-3-3 proteins, thereby inhibiting transcription of FOXO
target genes. In addition, FOXO proteins lacking Akt phos-
phorylation sites prevent IGF-I inhibition of DEX induction of
MuRF1 (48). Nevertheless, significant questions remain as to
whether nuclear FOXO transcription factors alone are suffi-
cient to activate transcription of the MuRF1 gene. In the
present study, we provide a detailed analysis of the regulatory
elements governing MuRF1 induction following glucocorti-
coid treatment, with particular attention to the role of the GR
and specific FOXO transcription factors.
MATERIALS AND METHODS
Cell culture. HepG2 and C
2
C
12
cells were cultured in DMEM
supplemented with 10% FBS, nonessential amino acids, and antibi-
otics and grown at 37°C in 5% CO
2
.C
2
C
12
cells were cultured in
DMEM, supplemented with 10% FBS, nonessential amino acids, and
antibiotics, and grown at 37°C in 5% CO
2
.C
2
C
12
myoblasts were
differentiated to myotubes by switching confluent cells (usually
24 48 h postsplit) to DMEM supplemented with 2% charcoal dex-
tran-treated FBS, nonessential amino acids, and antibiotics and grown
for an additional 48 –96 h at 37°C in 5% CO
2
. All cell culture reagents
except charcoal dextran-treated FBS (Hyclone) were from Invitrogen.
Plasmids. The pSG5-GR construct was a kind gift from Dr. Stoney
Simons, the pcDNA3-FoxO1 construct was provided by Dr. Masahiko
Negishi, pCMV5-cMyc-FoxO3a was provided by Dr. Dominic Accili,
and pcDNA3-FoxO4 was provided by Dr. Karen Arden. The
pcDNA3.1-GRwt construct contains the full-length mouse GR cDNA,
and the pcDNA3.1-GR
dim
construct was derived from pcDNA3.1-
GRwt by overlap PCR introducing the A458T point mutation and a
novel BsrGI restriction site, similar to the strategy used to create the
same point mutation in the GR
dim
mouse targeting vector described
previously (38). The mouse MuRF1 proximal promoter region was
obtained by PCR from bacteria artificial chromosome (BAC) clones
RP23-40E12 (Children’s Hospital Oakland Research Institute). BAC
DNA was isolated from bacterial cultures using the BACMAX DNA
purification kit from Epicentre according to the manufacturer’s pro-
tocol. Primers were designed to amplify approximately 5,000,
2,000, 1,000, and 500 bp from near the transcription start site.
The primer sequences used to generate the MuRF1 promoter frag-
ments are MuRF1-Pro5000-F 5-GGA CAG TGC ATC ATG ACC
CAG-3, MuRF1-Pro2000-F 5-CCA GAA CTA CAC CAG AAA CTC-
3, MuRF1-Pro1000-F 5-GGA GCT GGG AAT ATA GAC TTG-3,
MuRF1-Pro500-F 5-CCT TAG AGC TGT TCA GAA TCC AG-3, and
MuRF-Pro-R 5-CAC TCG GAT CCT CTT TGT CTT C-3. PCR was
performed using TaqPlus Long (Strategene). The resulting PCR prod-
ucts were then subcloned into the pGEMT-EZ vector (Promega) and
sequenced to confirm that the correct amplicon had been obtained.
The promoter fragments were then digested out of the pGEMT-EZ
vector with EcoRI, blunted, and cloned into the SmaI site of pGL3-
Basic vector (Promega), resulting in fusion with the firefly luciferase
reporter gene. The recombinant plasmids were then subjected to
restriction digest analysis and sequenced to confirm correct orienta-
tion. MuRF1 promoter fragments, fused to the secreted alkaline
phosphatase (SEAP) reporter gene, were constructed by digesting the
pGEMT-EZ recombinant plasmids with EcoRI and subcloning the
MuRF1 fragments into the EcoRI site of the pSEAP-Basic vector.
These constructs were then subjected to restriction digestion analysis
and sequenced to confirm correct orientation. Oligonucleotides cor-
responding to the predicted FOXO binding site [FOXO binding
element (FBE)] and the GRE in the MuRF1 promoter, as well as a
string of six diaminofluorescein (Daf-16) binding elements (6X-
DBE), were designed and ordered from Invitrogen. The oligo se-
quences are MuRF1-FBE-F 5-CTA GTT CTT GTT TAC GAC C-3,
MuRF1-FBE-R 5-CTA GGG TCG TAA ACA AGA A-3, MuRF1-
GRE-F 5-CTA GGC TCT GAA CAG TCT GTT CTT GTT-3, MuRF1-
GRE-R 5-CTA GAA CAA GAA CAG ACT GTT CAG AGC-3,
6X-DBE-F 5-CTA GAA GTA AAC AAC TAT GTA AAC AAC TAT
AAG TAA ACA ACT ATG TAA ACA ACT ATA AGT AAA CAA CTA
TGT AAA CAA GAT C-3, and 6X-DBE-R 5-CTA GGA TCT TGT
TTA CAT AGT TGT TTA CTT ATA GTT GTT TAC ATA GTT GTT
TAC TTA TAG TTG TTT ACA TAG TTG TTT ACT T-3. The
complementary oligo sequences were annealed by mixing and heating
to 95°C and slowly cooling to 25°C over a 35-min period using a
thermocycler. The annealed oligos were then ligated into the SpeI site
of the thymidine kinase (TK)-Luc vector (kindly provided by Dr.
Ronald Evans). The resulting recombinant plasmids were then restric-
tion digested to confirm the presence of an insert and sequenced to
determine the number and orientation of the concatemerized oligos.
Site-directed mutagenesis of the FBE and GRE in the pGL3- and
pSEAP-MuRF1-Pro500 constructs was performed essentially as de-
scribed in the site-directed mutagenesis kit protocol from Strategene.
The primers used to mutate the FBE and GRE are: MuRF1Pro-GRE-
Mut-F 5-CCT GGC TCT GGT CAG TCT GAC CTT GTT TAC G-3,
MuRF1Pro-GRE-Mut-R 5-CGT AAA CAA GGT CAG ACT GAC
CAG AGC CAG G-3, MuRF1Pro-FBE-Mut-F 5-CTG TTC TTG
GTG ACG ACC CCC-3, and MuRF1Pro-FBE-Mut-R 5-GGG GGT
CGT CAC CAA GAA CAG-3. The resulting clones were sequenced to
confirm that the correct mutation had been obtained. The mutated
nucleotides are underlined.
Cell culture reporter gene and Northern blot assays. One hundred
twenty-five to 150 10
3
HepG2 cells/well were plated into 12-well
plates and cultured for 24 h or until an approximate confluency of
30 40% was reached. Using FuGene 6 (Roche), 1 g of total
DNA/well was transiently transfected [including 0.250 g/well of the
E786 MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
indicated reporter construct, 0.070 g/well of SV40-Renilla luciferase
(Promega), 0.250 g/well GR expression vector (i.e., pSG5-GR,
pcDNA3.1-GRwt, or pcDNA3.1- GR
dim
), 0.125 g/well FOXO ex-
pression vector, i.e., pcDNA3-FoxO1, pCMV5-cMyc-FoxO3a, or
pcDNA3-FOXO4, and pBluescript as filler DNA] for 12–16 h. Cells
were then treated for 18 –24 h with either vehicle, 1 M DEX (from
a 1-mM stock in ethanol; Sigma), 20 ng/ml IGF-I (Sigma), or both as
indicated. Dual-luciferase reporter assays were performed in HepG2
cells essentially as described previously (7). Briefly, transfected cells
were lysed with 1X passive lysis buffer (Promega), scraped, trans-
ferred to microfuge tubes, and centrifuged at high speed for 5 min to
clear cellular debris, and then 25 l of supernatant from each tube
(corresponding to each well) was transferred to a 96-well plate. Both
working firefly buffer (25 mM glycylglycine, pH 8.0, 5 mM K
2
HPO
4
,
4 mM EGTA, 15 mM MgSO
4
, 4 mM ATP, 1.25 mM DTT, 0.1 mM
CoA, 80 MD-luciferin) and working Renilla buffer (1.1 M NaCl, 2.2
mM Na
2
EDTA, 0.22 M K
2
HPO
4
, pH 5.1, 0.5 mg/ml BSA, 1.5 mM
NaN
3
, and 1.5 M coelenterazine) were prepared fresh prior to each
assay. D-luciferin was purchased from ICN. Five milligrams of lucife-
rin was dissolved in 18 ml of 25 mM glycylglycine (pH 8.0), aliquoted,
and stored at 80°C. Coelenterazine (Promega) was dissolved in EtOH
to a final concentration of 3 mM. All other chemicals were purchased
from Sigma. Following treatments, cells were lysed and supernatants
analyzed for luciferase activity using a Beckman Coulter LD 400 lumi-
nometer programmed to dispense 100 l of working firefly buffer with a
1-s delay and a 10-s integration, followed by injection of 100 lof
working Renilla buffer with a 2-s delay and a 10-s integration. Firefly
luciferase activities were normalized to Renilla luciferase activity to
correct for variations in transfection efficiency.
For C
2
C
12
transfection experiments, 125–150 10
3
cells/well
were plated into 12-well plates and cultured as described above for
24 h. Using FuGene 6 (Roche), 1 g of total DNA/well was tran-
siently transfected (including 0.250 g/well of the indicated reporter
construct, 0.125 g/well of a TK--galactosidase construct, and
pBluescript as filler DNA) for 12–16 h. Cells were then switched to
DMEM containing 2% charcoal/dextran-treated FBS. Twenty-four
hours following the switch, the cells were treated with vehicle and
either 1 M DEX (from a 1-mM stock in ethanol; Sigma), 20 ng/ml
IGF-I (Sigma), or both as indicated. Medium from each well of the
transfected differentiated C
2
C
12
myotubes was sampled immediately
prior to ligand treatment and then every 24 h following ligand
treatment for 3 days. Fresh ligand was added to the differentiated
C
2
C
12
myotubes every 24 h. Ten to 15 microliters of each collected
medium was then processed and analyzed using the Great EscAPe
SEAP Detection Kit (BD Biosciences), following the instructions of
the manufacturer. At the end of the experiment, the C
2
C
12
myotubes
were lysed and -galactosidase activities determined and used to
correct for variations in SEAP activities resulting from variations in
transfection efficiencies.
For C
2
C
12
Northern blot experiments, cells were differentiated and
treated as above, and total RNA was isolated using using RNeasy
columns (Qiagen) according to the manufacturer’s instructions.
Northern analysis was conducted using random hexamer-labeled
MuRF1 and rpL8 cDNAs essentially as described previously (8).
Animal studies. The generation of GR
dim
mice was described
previously (38). The mice were backcrossed to Balb/c mice for more
than 20 generations. Homozygous mutants and wild-type controls
were obtained by intercrossing heterozygous GR
dim
mice. Animals
were housed individually in ventilated cages under specific pathogen-
free conditions with ad libitum access to food and water and a normal
dark-light cycle. For the 6- and 24-h DEX treatments, 10 mg/kg
water-soluble DEX (Sigma) was given by intraperitoneal injection,
and 50 mg/l DEX was subsequently provided in the drinking water.
For the 3- and 8-day DEX treatments, 50 mg/l water-soluble DEX was
provided in the drinking water and replenished every 2nd day. At each
time point, animals were weighed and euthanized, and spleens as well
as tibialis anterior and gastrocnemius muscles were dissected and
weighed. Muscles were frozen in liquid nitrogen until further process-
ing. Splenocytes were isolated by passing the freshly isolated spleen
through a 40-m nylon mesh, washed in PBS, and subsequently
treated with OptiLyse (Beckman Coulter) to remove the erythrocytes.
Total splenocyte numbers were determined by counting the cells
under the microscope with a Neubauer chamber. Total RNA from
treated wild-type and GR
dim
gastrocnemius muscle was isolated and
analyzed by Northern hybridization using MuRF1, FOXO1 and
FOXO3a, and rpl32 cDNA probes as described above. All animal
experimentation was conducted in accordance with standards of
human animal care and approved by the Lower Saxony state author-
ities (Niedersa¨chisches Landesamt fu¨r Verbraucherschutz und Leb-
ensmittelsicherheit, Braunschweig, Germany).
Chromatin immunoprecipitation. Chromatin immunoprecipitations
(ChIP) were performed as modified by Metivier et al. (30) and Nelson
et al. (35). Chemicals were obtained from Sigma unless stated other-
wise. C
2
C
12
cells were grown to confluence in 100-mm tissue culture
plates in DMEM supplemented with 10% FBS. Cells were then
shifted to 2% FBS and allowed to differentiate into myotubes for 4
days. One plate per time point for the 1-M DEX time course
treatment or four sets of plates in quadruplicate were treated with 1
M DEX, 20 ng/ml R3-IGF-I, or a combination of 1 M DEX and 20
ng/ml R3-IGF-I for 15 or 30 min. Untreated cells were used as a
control. At the indicated times, medium was removed and cells were
incubated with 1% formaldehyde in PBS for 10 min at 37°C. Form-
aldehyde cross-linking was quenched by adding glycine to a final
concentration of 125 mM and incubating for an additional 10 min at
room temperature. Cells were washed twice in PBS, scraped in 10
mM TrisHCl, pH 8.0, 150 mM NaCl, and 1 mM EDTA, and
collected by centrifugation at 3,000 gfor 5 min. Cell pellets were
resuspended in 400 l of 1% SDS, 10 mM EDTA, 50 mM TrisHCl,
pH 8.0, 10 mM -glycerophosphate, 1 mM sodium orthovanadate,
and protease inhibitor cocktail (Roche). Cells were sonicated twice for
30 s each on setting “3” and “pulsed” using a Branson microtip
sonicator. The resulting lysates were then centrifuged at full speed in
an Eppendorf microfuge for 10 min. Supernatants were diluted to 10
ml with ChIP dilution solution (0.01% SDS, 1.1% Triton X-100, 1.2
mM EDTA, 16.7 mM TrisHCl, pH 8.0, 167 mM NaCl, protease
inhibitor cocktail, 10 mM -glycerophosphate, and 1 mM sodium
vanadate). The indicated antibody [anti-HA tag IgG HA probe (Y-11),
anti-FOXO1 IgG (H-128), and anti-glucocorticoid receptor (M-20)
were obtained from Santa Cruz Biotechnology] was added to the
diluted chromatin at a concentration of 1 g/ml, and samples were
incubated overnight at 4°C with tumbling. Fifty microliters of protein
A (50% slurry) preabsorbed with sheared salmon sperm DNA was
added and incubated with tumbling at 4°C for 2 h. Immunoprecipita-
tions were transferred to Bio-Rad minicolumns (prerinsed with 500 l
of ChIP dilution solution), with an additional 500-l dilution solution
to rinse the tubes and ensure complete transfer of beads to the
minicolumns. Columns were then washed twice each with 1 ml TSEI
(0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM TrisHCl, pH
8.0, 150 mM NaCl), TSEII (0.1% SDS, 1% Triton X-100, 2 mM
EDTA, 20 mM TrisHCl, pH 8.0, 500 mM NaCl), and TSEIII (1 mM
EDTA, 10 mM TrisHCl, pH 8.0, 1% NP-40, 1% sodium deoxy-
cholate, 0.25 M LiCl) followed by three washes with Wash Buffer IV
[10 mM Tris (pH 8.0), 1 mM EDTA]. Washed beads were transferred
to new tubes with 400 l of Wash Buffer IV, adding an additional 500
l Wash Buffer IV to the columns to ensure that all beads were
transferred. The beads were then pelleted by centrifugation at 3,000 g
for 2 min. After the supernatant was removed by careful aspiration,
100 l of 10% Chelex-100 (Bio-Rad) was added to the beads,
followed by vortexing and incubation at 95°C for 10 min to reverse
the cross-linking. Two microliters of 20 U/ml proteinase K solution
(Invitrogen) was added, followed by vortexing and incubation at 55°C
for 30 min and heat inactivation at 95°C for 10 min. After centrifu-
gation at full speed in an Eppendorf microfuge for 2 min, the
supernatant was transferred to a new tube and combined with a second
E787MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
extraction of the Chelex beads with 100 l of water. Input (nonim-
munoprecipitated chromatin) was prepared by adding 100 l of 10%
Chelex directly to 100 l of the diluted chromatin extract and
processed as above. Quantitative PCR of input and immunoprecipi-
tated chromatin samples was performed using 2 l of DNA as
template and PerkinElmer 2X SYBR Green master mix on an ABI
7700. Cycling conditions were one cycle at 95°C for 10 min, followed
by 40 cycles at 95°C for 10 s and 60°C for 60 s. Primers flanking the
putative mouse MuRF1 GRE/FBE region were forward 5-
TATCTGGCTCTCCCCTGAAC-3and reverse 5-CCTCAAAGATTTG-
GCCCTCT-3. Values for each time point and hormone treatment were
normalized to input values. For agarose gel analysis of PCR products,
reactions were stopped at 30 cycles, and samples from each treatment
group were pooled and run on a 2% agarose gel containing ethidium
bromide.
RESULTS
The MuRF1 promoter contains a conserved near-perfect
palindromic glucocorticoid response element. Since MuRF1 is
known to be induced by synthetic glucocorticoids, as well as in
several catabolic conditions associated with elevated endoge-
nous glucocorticoids, we sought to identify the key glucocor-
ticoid-responsive elements in the gene. We began by isolating
BAC clones encompassing the entire mouse MuRF1 transcrip-
tion unit and amplifying 5,000 bp upstream of the transcription
start site of the gene. Human, rat, and mouse promoter se-
quences were aligned to detect conserved sequences that may
be functionally relevant for MuRF1 expression. The MuRF1
(Fig. 1A) promoter shows a high degree of homology across
Fig. 1. Schematic of the muscle RING finger 1
(MuRF1) promoter and sequence alignment of the
proximal regulatory regions. A: promoter sequences
from mouse, rat, and human MuRF1 [5,000 base
pairs upstream of the transcription start site (1)
through the first exon] were downloaded from the
Ensembl database (www.ensembl.org) and aligned
using the ClustalW algorithm. Approximate positions
of potential transcription factor binding sites are in-
dicated in the schematics of the MuRF1 promoter at
top and/or boxed in the alignment at bottom. The
forkhead transcription factor class O (FOXO) fork-
head binding site (G/A)TAAA(T/C)AA (black
ovals), glucocorticoid response element (GRE) (A/
T)GAACANNNTGTTC(A/T) (hatched rectangle),
CCAAT/enhancer-binding protein (C/EBP) TT(G/
T)NGNAA ({), NF-B consensus binding sequence
GGG(G/A)N(C/T)(C/T)(C/T)CC (gray hexagons),
and muscle-specific E-box CANGTG (MyoD, etc.);
N represents any nucleotide. The sequence align-
ments for 400 base pairs upstream and 26 base
pairs downstream of the rat, mouse, and human
MuRF1 promoters are shown below the correspond-
ing promoter schematics. Identical sequences for the
indicated regions are highlighted in black. Arrow
indicates transcription start site. B: comparison of a
consensus GRE and perfect palindrome GRE and
putative GREs from the mouse (mMuRF1), rat
(rMuRF1), and human MuRF1 (hMuRF1) promoters.
Arrows indicate half-sites.
E788 MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
species for about 400 bp upstream of the transcription start site,
and there are additional sporadic pockets of homology far
upstream of the proximal promoter, 3of the transcription unit,
and within some introns (not shown). We searched for poten-
tial transcription factor binding sites by scanning the promoters
for published and well-established consensus sequences. Pre-
vious reports identified and characterized consensus Class O
forkhead (FOXO) binding sites in the promoter of the muscle
atrophy-related muscle atrophy F-box gene (42), and we found
multiple potential AT-rich FBEs in the MuRF1 promoter as
well, including one overlapping the consensus TATA box (Fig.
1A). Importantly, the MuRF1 promoter has a near-perfect
consensus GRE 200 bp upstream of the transcription start
site, supporting our hypothesis that this gene could be a direct
target of the ligand-activated GR. The potential GREs from the
human, mouse, and rat MuRF1 genes differ only by a single
nucleotide in the 3-bp spacer region, whose exact sequence is
not critical for GR binding (Fig. 1B). Interestingly, a consensus
FBE was found directly adjacent to the putative MuRF1 GRE,
raising the possibility that FOXO and the GR function together
to regulate the MuRF1 promoter. In addition to FOXO and GR
binding sites, we detected a cluster of potential NF-B re-
sponse elements in the MuRF1 promoter, consistent with the
upregulation of this promoter by proinflammatory cytokines
and upon activation of NF-B (4). Several C/EBP sites were
also detected in the MuRF1 promoter, which is of interest
because C/EBPand -are upregulated by DEX in skeletal
muscle cells (57, 58). Finally, numerous conserved consensus
muscle-specific E-box sites were found in the promoter, as
expected given its highly selective cardiac and skeletal muscle
expression pattern.
The MuRF1 promoter is a direct target of activated GRs.
Since the mere presence of consensus transcription factor
binding sites does not guarantee their functional importance,
MuRF1 promoter fragments, or the predicted GRE and FBE in
a minimal heterologous promoter, were placed in front of the
firefly luciferase reporter and transiently transfected into
HepG2 cells along with expression vectors for the murine GR
and/or FOXO family members. For these initial studies,
HepG2 cells were chosen for their ease of transfection and their
previous use in transfection studies of gluconeogenic gene
promoters that are also regulated by both GR and FOXO
family members (23, 24, 52). Consistent with the presence of
the consensus GRE within 500 bp of the transcription start site,
DEX was able to induce all MuRF1 promoter constructs
containing between 500 and 5,000 bp (Fig. 2A). Indeed, the
isolated MuRF1 GRE is sufficient to support potent DEX-
induced transcription either alone (14-fold) or as a multimer
(19- to 45-fold) (Fig. 2B).
FOXO transcription factors differentially activate the
MuRF1 promoter. Next, we tested whether the native MuRF1
promoter would respond to cotransfected FOXO1, FOXO3a, or
FOXO4 expression vectors, since these proteins are candidates
for intermediate DEX-inducible regulatory factors. As shown
in Fig. 3A, FOXO factors only marginally induce the proximal
MuRF1 promoter (2- to 3-fold). As with the GRE, we tested
whether the isolated FOXO binding site (FBE) would respond
to cotransfected FOXO1, FOXO3a, or FOXO4 expression
vectors; in this context, only FOXO3a and FOXO4 induced
transcription of the reporter containing a single FBE (Fig. 3B).
However, FOXO1 is able to activate multimerized DBE or the
MuRF1 FBE, suggesting that this FOXO family member is
most active as a multimeric complex rather than a monomer
(Fig. 3C).
The MuRF1 promoter is synergistically activated by FOXO1
and GR. Remarkably, however, FOXO1 showed strong syner-
gistic activation with the GR on the 500-bp MuRF1 promoter
(Fig. 4A). This potent synergy was consistently observed (20-
to 40-fold) over multiple experiments and on all promoter
fragments tested 5,000 bp from the transcription start site.
FOXO3a and FOXO4 did not synergize with the GR; indeed,
expression of FOXO3a somewhat inhibits GR activation of the
MuRF1 promoter. The GR-FOXO1 synergy is also reflected in
dose-response experiments, where this combination supports
strong activation of the MuRF1 promoter with as little as 1 nM
DEX in the culture medium (Fig. 4B). Both the GRE and the
adjacent FBE play an important role in the observed GR-
FOXO synergy. DEX induction of the 500-bp MuRF1 pro-
moter is nearly abolished when the GRE is mutated and is
completely abolished when the GRE is intact but the adjacent
FOXO site is mutated (Fig. 4, Cand D).
DEX induction of MuRF1 expression is inhibited in GR
dim
mice that express a dimerization-deficient GR. The functional
significance of the near-perfect GRE in the MuRF1 promoter
was tested in vivo through the use of the GR
dim
mouse strain
that expresses a GR with a point mutation in the DNA binding
domain that prevents binding of the GR to classical palin-
Fig. 2. Dexamethasone (DEX)-activated glucocorticoid receptor (GR) induces
the MuRF1 proximal promoter. A: HepG2 cells were transfected with lucif-
erase reporter constructs (Luc) containing varying lengths of the MuRF1
(pGL3-MuRF1) promoter, an SV40-Renilla luciferase reporter construct, and
a mouse GR expression vector (pSG5-GR). Cells were treated with or without
1M DEX for 24 h. Luciferase activities in cell extracts were normalized to
Renilla luciferase activity to control for transfection efficiency. Numbers on
the y-axis indicate distance from the transcription start site fused to the reporter
pGL3-Basic, a promoterless LUC vector. B: the isolated MuRF1 GRE supports
DEX-mediated transcriptional activation of a heterologous promoter. The DEX
regulation of the thymidine kinase (TK) promoter was tested with 0 (control),
1 (1XGRE), 3 (3XGRE), and 4 (4XGRE) inserted GREs. The arrows below the
indicated GRE constructs labeled on the y-axis depict the orientation of each
individual GRE oligonucleotide. In Aand B, fold induction was obtained by
dividing values from DEX-treated samples by the mean of values from
matched untreated samples. Each condition was done in triplicate, and error
bars reflect SD.
E789MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
dromic GREs (38). This mutant GR still retains the ability to be
tethered to target genes via protein-protein interactions such as
the NF-B or AP-1 transcription factor complexes. Both wild-
type and GR
dim
mice were treated with water-soluble DEX
either by injection (6- and 24-h time points) or in the drinking
water (24-h, 3-day, and 8-day time points). The experiment
was terminated at 8 days of treatment due to the severe weight
loss observed in the wild-type mice, which was not observed in
the GR
dim
mice (P0.001). As shown in Fig. 5A, despite the
inhibited overall body weight decline in GR
dim
vs. wild-type
mice in response to DEX, the degree of atrophy induced in the
tibilialis anterior and gastrocnemius muscles was not statisti-
cally different in the two strains. By contrast, DEX-induced
splenocyte apoptosis was strongly inhibited in GR
dim
mice
(P0.001).
We next examined MuRF1 expression as well as FOXO1
and FOXO3a expression in GR
dim
vs. GR
/
mice (Fig. 5B).
MuRF1 expression is clearly upregulated by 6 h after DEX
injection and continues to increase 3 days with continuous
DEX treatment in wild-type mice. MuRF1 induction is
strongly inhibited in GR
dim
mice, which is particularly appar-
ent at the earliest time points. After 8 days of DEX treatment,
however, MuRF1 expression declines in wild-type mice until it
is essentially equal to its induced expression in GR
dim
animals.
In addition, FOXO1 and FOXO3a are also rapidly induced by
DEX in wild-type mice, in agreement with a previous study
(9), and expression is reduced in GR
dim
mice at all time points
tested. We then tested the ability of the homodimerization
mutant GR expressed in GR
dim
mice to upregulate the proximal
MuRF1 promoter, again using transient transfection assays
(Fig. 5C). The homodimerization mutant GR lost the ability to
induce the MuRF1 promoter on its own, as expected, and
showed a strongly reduced, but not completely abolished,
ability to augment FOXO1 induction of the promoter. These
transfection results may at least partially explain the residual
DEX-induced expression of MuRF1 in GR
dim
mice.
The GR and FOXO1 bind to the endogenous MuRF1 pro-
moter in C
2
C
12
myotubes. Given the strong dependence on GR
homodimerization in vivo for full MuRF1 gene induction, we
next sought to determine the role of the GRE and FOXO1 sites
in cultured skeletal myotubes. As we observed in HepG2 cells,
the 500-bp MuRF1 promoter is also DEX inducible in differ-
entiated C
2
C
12
mouse skeletal myotubes (Fig. 6A). DEX in-
duction of the promoter is nearly abolished when the GRE is
mutated, and the adjacent FOXO site is intact and completely
abolished when the GRE is intact but the adjacent FOXO site
is mutated (Fig. 6B). Furthermore, addition of 20 ng/ml IGF-I
strongly inhibits DEX induction of the endogenous MuRF1
gene, as has been reported previously (Fig. 6C) (41, 48), and
IGF-I likewise inhibits DEX induction of the MuRF1 promoter
(Fig. 6D). Therefore, the 500-bp proximal promoter of MuRF1
contains the critical elements necessary to respond positively to
glucocorticoids (via synergy with a forkhead site) and nega-
Fig. 3. FOXO transcription factor induction of the MuRF1 promoter. A: HepG2 cells were transfected with 500-base pair MuRF1-promoter-LUC constructs,
SV40-Renilla luciferase reporter construct, and expression vectors for murine FoxO1, FoxO3A, or FoxO4 (pcDNA3-FoxO1, pcDNA3-FoxO3A, or pcDNA3-
FoxO4) or pcDNA3 alone (dash). Luciferase activities in cell extracts were normalized to Renilla luciferase activity to control for variations in transfection
efficiency. Each point was done in triplicate; error bars reflect SD. B: the isolated MuRF1 FOXO binding element (FBE) supports FOXO3a- and FOXO4- but
not FOXO1-mediated transcriptional activation of a heterologous promoter. FOXO regulation of the TK promoter was tested by inserting a single FBE and
cotransfecting HepG2 cells with the control pcDNA3 vector alone (dash) or each FOXO expression vector as in A.C: concatamerized FOXO binding elements
from the MuRF1 promoter (FBE-4X) and Daf-16 binding elements (DBE-6X) support transcriptional activation of the TK promoter by FOXO1. Open bars,
normalized luciferase values from the cells transfected with the TK-Luc-4XFBE and TK-Luc-6XDBE in the absence of exogenous FOXO1 expression; black
bars, normalized luciferase values from the TK-Luc-4XFBE- and TK-Luc-6XDBE-transfected cells in the presence of exogenous FOXO1 expression. Data were
processed as in Aand B.
E790 MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
tively to IGF-I in a manner identical to the endogenous gene in
differentiated myotubes.
To determine whether FOXO1 and GR can directly occupy
the MuRF1 promoter in vivo, we established a modified ChIP
protocol suitable for use with differentiated C
2
C
12
myotubes.
C
2
C
12
myoblasts were plated on 100-mm dishes and cultured
until confluent and then differentiated for 4 days in reduced
serum medium. Myotubes were then treated with vehicle, 1
M DEX, 20 ng/ml IGF-I, or 1 M DEX plus 20 ng/ml IGF-I
for the indicated times. Sonicated chromatin from fixed cells
was analyzed with control IgG, an anti-FOXO1 antibody, or
two anti-GR antibodies. Both GR and FOXO1 are present on
the endogenous MuRF1 promoter in untreated C
2
C
12
myotubes
(Fig. 7A), and binding of each factor is enhanced by DEX
treatment as soon as 15 min after ligand addition (Fig. 7, Band
C), especially the GR. GR binding to the MuRF1 promoter is
unaffected by IGF-I (Fig. 7B), although the DEX induction of
the endogenous gene or the transfected MuRF1 promoter is
strongly inhibited. However, IGF-I addition rapidly induces the
loss of FOXO1 from the MuRF1 promoter in the presence or
absence of DEX (Fig. 7C). We also attempted to detect
FOXO3a binding to the MuRF1 promoter by ChIP as well;
only one of two commercially available antibodies we tested
(Upstate vs. Santa Cruz) gave a weak positive signal, and the
binding pattern was highly similar to FOXO1 (not shown).
DISCUSSION
Although it is known that MuRF1 expression increases
under numerous atrophy-inducing conditions (11), the mecha-
nism of transcriptional regulation of this gene is poorly under-
stood. The present study provides new insights into how the
GR and the FOXO family of transcription factors regulate the
transcription of the MuRF1 gene. Specifically, we demonstrate
here that 1) the MuRF1 proximal promoter is directly activated
by glucocorticoids via a conserved GRE that depends strongly
on GR homodimerization for full activation, at least in the
earliest time points after DEX treatment, 2) FOXO1 and GR
synergistically and specifically induce the MuRF1 proximal
promoter, and 3) IGF-I inhibition of DEX-induced MuRF1
expression correlates with the loss of FOXO1 binding to the
endogenous MuRF1 promoter. Importantly, these data also
reveal that not all FOXO family members equally activate the
FOXO binding motif in the MuRF1 promoter.
The presence of an essentially perfect palindromic GRE in
the MuRF1 proximal promoter likely explains the strong in-
duction of the gene by exogenous synthetic glucocorticoids (3,
48) and how the gene is induced under a variety of catabolic
conditions associated with increased endogenous corticoste-
roid levels (25, 53). Each half-site of the MuRF1 GRE is
perfectly conserved between the human, rat, and mouse genes,
with the only difference found in the spacer region that,
nevertheless, still retains the three-base pair length necessary
for GR homodimer binding (43). It is surprisingly rare to find
such a perfect GRE in an endogenous glucocorticoid target
gene promoter, although the specific sequences of the diver-
gent half-sites found in most direct GR target genes are
strongly conserved across species (45, 46). Most GREs, such
as the pair found in the well-studied phosphoenolpyruvate
Fig. 4. DEX-activated GR synergizes with FOXO1 to potently induce the MuRF1 promoter. A: HepG2 cells were transfected with the pGL3-MuRF1 promoter
(500) reporter construct, SV40-Renilla luciferase reporter construct, pSG5-GR, and/or pcDNA3-FOXO1, pcDNA3-FoxO3A, or pcDNA3-FOXO4. Cells were
treated with or without 1 M DEX for 24 h. Firefly luciferase activities in cell extracts were normalized to Renilla luciferase activity to control for variations
in transfection efficiency. Fold induction was obtained by dividing values from DEX-treated samples by the mean of values from matched untreated samples.
Each point was done in triplicate, and errors reflect SD. B: HepG2 cells were transfected with pGL3-MuRF1 promoter (500), SV40-Renilla, and pBluescript
as filler DNA (control) and either GR (GR) or GR and FoxO1 (GR FoxO1) expression vectors. Twenty-four hours posttransfection, cells were treated with
the indicated DEX concentration and incubated overnight. Each point was done in triplicate. Cand D: mutation of either the GRE or the FBE is sufficient to
abolish DEX-induced MuRF1 promoter activity. HepG2 cells were transfected with either the wild-type MuRF1 promoter construct (500) or MuRF1 promoter
constructs that have either the GRE mutated (GRE-Mut) or the FBE mutated (FBE-Mut) as shown in Cin combination with expression vectors for FoxO1 and/or
GR as indicated. The cells where then treated and assayed for luciferase activity as in A.
E791MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
carboxykinase (PEPCK) gene in the liver (18) or the first-
described GREs in the mouse mammary tumor virus long-
terminal repeat (37), have multiple base pair changes compared
with the perfect palindrome consensus GRE that is present in
the MuRF1 promoter. Previous studies on the estrogen-regu-
lated family of vitellogenin genes in the frog liver demonstrate
that a single perfect estrogen response element supports essen-
tially the same fold induction by estradiol as a pair of imperfect
palindromic elements (29). The physiological relevance of
having such a well-conserved consensus GRE in the MuRF1
promoter is an interesting and important question.
Analysis of MuRF1 induction by DEX in GR
dim
mice
revealed a critical role for GR homodimerization, which is
most evident at the earliest time points examined in these
experiments. Interestingly, despite the clear overall reduction
in MuRF1 expression, as well as FOXO1 and FOXO3a, the
degree of muscle atrophy in GR
dim
mice after 8 days of DEX
treatment was exactly the same in the mutant as in wild-type
mice. Consequently, the residual level of MuRF1 expression in
GR
dim
mice may well be enough to support full DEX-induced
atrophy. The rapid rise and gradual decline of MuRF1 expres-
sion we observed in DEX-treated Balb/c mice is in agreement
with experiments in rats dosed with the synthetic glucocorti-
coid prednisolone (1). Concerning the mechanism of residual
MuRF1 induction in GR
dim
mice, it is conspicuous that co-
transfection of FOXO1 with the homodimerization mutant GR
0
5
10
15
20
25
30
35
40
Control GR FOXO1 GR + GRdim GRdim +
FOXO1 FOXO1
Fold induction (+DEX/-DEX)
35
40
45
50
55
Tibialis anterior weight (mg)
0GRdim
1
2
4
8
16
32
64
128
256
GRdim
70
90
110
Gastrocnemius weight (mg)
0GRdim
-20
-15
-10
-5
0
5
Reduction in body weight (%)
GR+/+ GRdim
GR+/+
GR+/+ GR+/+
-DEX
+DEX
-DEX
+DEX
-DEX
+DEX
A
B
C
GR+/+ GRdim
Untreated 3d DEX 8d DEX
MuRF1
FOXO1
FOXO3a
rpL32
GR+/+ GRdim GR+/+ GRdim
GR+/+ GRdim
Untreated 6 hr DEX 24 hr DEX
GR+/+ GRdim GR+/+ GRdim
MuRF1
FOXO1
FOXO3a
rpL32
Splenocytenumber (x106)
Fig. 5. DEX-induced MuRF1 expression is inhibited in mutant GR homodimerization mutant (GR
dim
) mice vs. wild-type GR
/
mice. A:GR
/
and GR
dim
mice were treated with DEX in their drinking water for 8 days, weighed (top left), and killed for determination of splenocyte number (top right) as well as
gastrocnemius (bottom left) and tiblialis anterior (bottom right) weights. Open bars, vehicle treated; black bars, DEX treated. GR
/
,n8; GR
dim
,n7.
B: Northern analysis of MuRF1, FOXO1, and FOXO3a expression in gasctocnemius muscle from DEX-treated GR
/
and GR
dim
mice. B,top: control, 6-h-,
and 24-h-treated mice. B,bottom: control, 3-day-, and 8-day-treated mice. Only results from 4 of the 8-day-treated GR
/
and GR
dim
mice are presented. rpL32
expression is shown as a loading control below each set of Northerns. Bars represent means SE. C: effect of wild-type GR and the homodimerization mutant
GR on DEX induction of the MuRF1 promoter in a transient transfection assay. HepG2 cells were transfected with the pGL3-MuRF1 promoter (500) reporter
construct, SV40-Renilla luciferase reporter construct (control), pcDNA3.1-GRwt (GR) or pcDNA3.1-GR
dim
(GR
dim
), and/or pcDNA3-FoxO1 (FOXO1). Cells
were treated with or without 1 M DEX for 24 h. Firefly luciferase activities in cell extracts were normalized to Renilla luciferase activity to control for variations
in transfection efficiency. Fold induction was obtained by dividing values from DEX-treated samples by the mean of values from matched untreated samples.
Each point was done in triplicate, and errors reflect SD.
E792 MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
supports at least modest induction of the MuRF1 promoter in
transient transfection assays, whereas the induction without
FOXO1 is completely lost (Fig. 5). This is in line with the
previous observation that the GR
dim
receptor on its own fails to
bind to a palindromic GRE. Nevertheless, it is conceivable that
FOXO1, when present together, still tethers the homodimer-
ization mutant GR to the composite GRE-FBE motif, since
protein-protein interactions with other transcription factors are
not compromised by the GR
dim
mutation. Regardless, it will be
important to identify DEX-responsive genes that are less af-
fected than MuRF1 and FOXOs in GR
dim
mice that may
ultimately be more important for the loss of muscle mass in
response to DEX. For example, activation of the N-end rule
ubiquitin pathway was recently implicated in MyoD degrada-
tion in response to DEX in C
2
C
12
cells (49). In addition,
transgenic overexpression of MuRF1 was not sufficient to
induce atrophy but led to altered levels of enzymes involved in
carbohydrate metabolism (16). Thus, consideration of these
and other studies, and our present results with GR
dim
mice,
may force a reevaluation of the role of MuRF1 (and possibly
even FOXO1 and FOXO3a) in the reduction of muscle mass in
catabolic states, although the protein certainly may still play a
significant role in structural protein degradation in denervation-
induced atrophy. Further analysis of the GR
dim
model may
allow for a genetic dissection of the induction of atrophy vs.
metabolic effects of glucocorticoids in this important GR target
tissue.
Several recent papers have implicated FOXO family tran-
scription factors as being important for the upregulation of
gene expression during skeletal muscle atrophy, including after
DEX treatment or nutrient starvation in C
2
C
12
cells. For
example, Stitt et al. (48) reported that activated FOXO1 is
necessary but not sufficient to activate the MuRF1 gene in
cultured myotubes; however, the effects of FOXO3a or
Fig. 6. DEX induces the MuRF1 promoter in differentiated C
2
C
12
myotubes.A:C
2
C
12
myoblasts were transfected with a reporter construct containing 500 base
pairs of the MuRF1 promoter fused to the secreted alkaline phosphatase (SEAP) gene. The myoblasts were then differentiated by switching to low-serum medium
followed by treatment with 1 M DEX over a period of 3 days. The medium was sampled every 24 h to measure for SEAP activity. Conditions were done in
triplicate and SEAP numbers normalized with -galactosidase to correct for variations in transfection efficiency. Each point was done in triplicate, and errors
reflect SD. B: mutation of either the GRE or the FBE is sufficient to abolish DEX-induced MuRF1 promoter activity in C
2
C
12
cells. C
2
C
12
myoblasts were
transfected with either the wild-type MuRF1 promoter construct (500) or MuRF1 promoter constructs that have either GRE-Mut or FBE-Mut SEAP constructs,
differentiated, and treated with 1 M DEX over a period of 3 days. The medium was sampled every 24 h to measure for SEAP activity as in A. Each time point
was done in triplicate and normalized with -galactosidase to correct for variations in transfection efficiency. C: Northern blot analysis of C
2
C
12
cells
differentiated for 48 h and then treated with DEX (10 M), IGF-I (20 ng/ml), or DEX (10 M) IGF-I (20 ng/ml) for 24, 48, and 72 h. Ligand was refreshed
every 24 h. D: IGF-I potently inhibits DEX-induced activation of the MuRF1 promoter. C
2
C
12
myotubes were treated for 24 h with either 1 M DEX, 20 ng/ml
IGF-I, or 1 M DEX and 20 ng/ml IGF-I followed by sampling of the medium for SEAP activity, as described in A. Fold induction was obtained by dividing
values from DEX-treated samples by the mean of values from matched untreated samples. Each point was done in triplicate, and errors reflect SD.
E793MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
FOXO4 were not reported. Interestingly, selective transgenic
overexpression of FOXO1 in skeletal muscle produced mice
with significantly smaller muscles than wild-type litter mates
without apparent upregulation of MuRF1 (19). The lack of
MuRF1 induction in these instances is not surprising given our
present findings. Assuming that FOXO1 was the only tran-
scription factor overexpressed in these transgenic mice, and
that circulating corticosteroid levels are normal, then MuRF1
would not necessarily be upregulated. The reduction in muscle
mass observed in these mice may be due to the activation of
other atrophy-inducing pathways. For example, overexpression
of FOXO1 leads to an upregulation of 4E-BP1 and inhibition
of mTOR signaling, resulting in a decrease in protein
synthesis (47).
There is important precedence for the collaborative role of
FOXO1 on GR target genes from studies of inversely regulated
insulin and glucocorticoid-responsive genes in the liver. The
IGF-binding protein-1 (10) and glucose-6-phosphatase pro-
moters contain GREs with directly adjacent and even overlap-
ping FOXO binding sites, and the PEPCK (13) and pyruvate
dehydrogenase kinase-4 (24) genes have FOXO sites in rela-
tively close proximity to their GREs. In each case, integrity of
the FOXO site is important for maximal glucocorticoid respon-
siveness.
In addition, like MuRF1, the ability of FOXO1 and FOXO3a
to have differential effects on a single promoter has recently
been demonstrated by Onuma et al. (36) for the glucose-6-
phosphatase promoter. It is intriguing that key genes involved
in gluconeogenesis have clustered functional GREs and FOXO
binding sites in critical glucocorticoid and insulin-sensitive
enhancers reminiscent of our findings with the muscle-specific
MuRF1 gene promoter. It is tempting to speculate that if
indeed one role of MuRF1, as an E3 ubiquitin ligase, is to
target specific skeletal muscle proteins for degradation, then a
gene involved in providing a source of amino acid substrates
for the liver to use to make glucose de novo might logically be
under similar regulatory control as important gluconeogenic
enzymes. The common regulatory strategy of MuRF1 expres-
sion with liver gluconeogenic enzymes also supports the notion
of a potential prominent role for MuRF1 in skeletal muscle
glucose metabolism.
The role of FOXO1 as a key component of a composite
glucocorticoid-responsive unit in MuRF1 gene regulation is
further emphasized by our ChIP data. IGF-I not only virtually
eliminates DEX upregulation of the 500-bp MuRF1 promoter
and the endogenous MuRF1 gene (see Fig. 7) but also rapidly
clears the MuRF1 promoter of FOXO1 binding without affect-
ing GR binding. On the PEPCK enhancer in rat liver H4IIE
cells, insulin treatment causes a rapid loss of FOXO binding in
control and DEX-treated cells, with significant reduction al-
ready apparent by as little as 3 min after insulin addition (13).
In this case, GR binding was reduced to 40% compared with
the DEX only-induced level after 30 min of treatment, which
we did not observe upon IGF-I addition to C
2
C
12
cells. Nev-
ertheless, the most impressive change in promoter occupancy
examined in both our experiments and the PEPCK gene ex-
periments was decreased FOXO1 binding. In addition to inhi-
bition of FOXO1 (and to a lesser extent GR) binding to the
PEPCK promoter in response to insulin treatment, dramatic
changes in histone posttranslational modifications that paral-
leled decreased transcription were observed (13). Whether
Fig. 7. GR and FOXO1 associate directly with the MuRF1 promoter in C
2
C
12
myotubes. A:C
2
C
12
myotubes were treated with 1 M DEX for 0 or 60 min
(1 10-cm plate/time point), and cross-linked chromatin was immunoprecipi-
tated with normal rabbit IgG, an anti-FOXO1 antibody, or one of two anti-GR
antibodies (P20 or M20). After reversal of cross-links, immunoprecipitated
MuRF1 promoter fragments were detected by PCR using primers flanking the
predicted GRE and FOXO sites in the mouse MuRF1 promoter, followed by
agarose gel electrophoresis. PCRs of a no-template control (NTC) and input
DNAs are shown at left.Band C:C
2
C
12
myotubes were treated with vehicle
(open bars), 1 M DEX, 20 ng/ml R3-IGF-I, or 1 M DEX 20 ng/ml
R3-IGF-I for 15 min (black bars) or 30 min (gray bars) before harvest. Four
plates for each treatment were processed in parallel as in A. Values (fold
enrichment) are expressed as the mean MuRF1 promoter copies immunopre-
cipitated with either the anti-GR antibody (B) or anti-FOXO1 antibody (C)
divided by that immunoprecipitated by a nonspecific antibody (anti-HA), as
determined by quantitative PCR using the same primer pairs as in A. Error bars
reflect SE.
E794 MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
these changes occur on the MuRF1 promoter in response to
DEX and/or IGF-I treatment remains an interesting question,
as is whether removal of FOXO1 is the key event triggering the
reversal of transcriptional activation and associated chromatin
modifications induced by DEX.
The exact nature of the synergy between the GR and FOXO1
is clearly an important area for further investigation because it
is also not well understood for liver gluconeogenic genes.
FOXO1 has been demonstrated to physically interact in solu-
tion with several steroid hormone receptors, such as the estro-
gen, progesterone, and androgen receptors (21, 26, 44, 59), but
not, to date, with the glucocorticoid receptor. The functional
significance of this interaction (either repressing or enhancing
receptor activity) varies greatly depending on the cell type and
target gene promoter context. For example, FOXO1 synergis-
tically activates the IGF-binding protein-1 promoter with
ligand-activated progesterone receptors in endometrial adeno-
carcinoma cells but not in endometrial fibroblasts (20).
FOXO1, FOXO3a, and FOXO4 all contain an LXXLL motif
that can interact with a ligand-induced pocket on the hormone
binding domain of steroid receptors (59). Since FOXO1 bind-
ing to the MuRF1 promoter is (at least modestly) enhanced by
DEX treatment of C
2
C
12
cells along with the strong recruit-
ment of GR (Fig. 7), a situation also observed on the PEPCK
and glucose-6-phosphatase genes (13, 51), these proteins may
be mutually bound and stabilized on the DEX-activated
MuRF1 promoter. Synergy may also be achieved via corecruit-
ment of transcriptional coactivators, such as histone acetyl-
transferases like CBP/p300, that have been shown to be inde-
pendently recruited by the liganded GR or FOXO1 in other
contexts (28, 34). Irrespective of the precise nature of the
GR-FOXO1 synergy, our results reveal that each factor is
required to create an optimal glucocorticoid-inducible MuRF1
promoter.
There are several important implications of our findings on
MuRF1 gene regulation. First, the activated GR may be an
important player in the control of muscle atrophy, but the
presence of nuclear FOXO1 is critical to achieve full induction
of the MuRF1 promoter and, quite possibly, multiple glucocor-
ticoid-regulated genes in skeletal muscle. Furthermore, the
glucocorticoid concentration needed to induce MuRF1 is sig-
nificantly reduced in the presence of FOXO1. A logical pre-
diction on the basis of these results is that conditions with both
elevated corticosteroids and active FOXO1 would lead to the
greatest degree of muscle wasting and altered metabolism. In
fact, diabetes mellitus presents such a condition (53). In several
models of diabetes, Akt is less active, potentially leading to
decreased inhibition of FOXO activity and/or increased nuclear
localization (55). Elevated nuclear FOXO1 would be available
to interact with the GR on the MuRF1 promoter to further
induce the gene above the level induced by corticosteroids
alone. Prolonged glucocorticoid exposure also leads to insulin
resistance (54), which might in turn lead to higher MuRF1
induction, accelerated muscle atrophy, and metabolic changes
as part of a vicious cycle. In contrast, the need for both nuclear
FOXO1 and activated GR to fully activate MuRF1 may be
protective and prevent unnecessary muscle breakdown. For
example, following intense exhaustive exercise, circulating
cortisol levels increase (6, 31), yet elevated cortisol is generally
not associated with an increase in the breakdown of contractile
proteins, nor is loss of muscle mass a consequence of endur-
ance training (33). Thus, the need for additional factors to fully
activate MuRF1 gene expression could protect the muscle from
breakdown under certain physiological conditions.
In summary, our results demonstrate a potent synergy be-
tween the GR and FOXO1 in transcriptional control of an
important gene linked to skeletal muscle atrophy and metabolic
control. Further understanding the molecular details of the
opposing effects of corticosteroids and insulin/IGF-I on gene
expression in skeletal muscle, the breadth of the GR-FOXO1
cooperation on muscle gene expression in general, and the very
nature of this robust synergy should have important ramifica-
tions for preventing inappropriate induction of genes involved
in muscle atrophy and altered metabolism in catabolic disease
states.
ACKNOWLEDGMENTS
We acknowledge Dr. Frank Gannon and members of the Gannon Labora-
tory at the European Molecular Biology Laboratory for generous support and
advice on the ChIP experiments.
GRANTS
This work was supported by a Kirchstein National Research Service Award
to D. S. Waddell and National Institute of Diabetes and Digestive and Kidney
Diseases Grant RO1-DK-075801 to J. D. Furlow and S. C. Bodine.
REFERENCES
1. Almon RR, DuBois DC, Yao Z, Hoffman EP, Ghimbovschi S, Jusko
WJ. Microarray analysis of the temporal response of skeletal muscle to
methylprednisolone: comparative analysis of two dosing regimens.
Physiol Genomics 30: 282–299, 2007.
2. Beato M, Chalepakis G, Schauer M, Slater EP. DNA regulatory
elements for steroid hormones. J Steroid Biochem 32: 737–747, 1989.
3. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA,
Poueymirou WT, Panaro FJ, Na E, Dharmarajan K, Pan ZQ, Valen-
zuela DM, DeChiara TM, Stitt TN, Yancopoulos GD, Glass DJ.
Identification of ubiquitin ligases required for skeletal muscle atrophy.
Science 294: 1704 –1708, 2001.
4. Cai D, Frantz JD, Tawa NE Jr, Melendez PA, Oh BC, Lidov HG,
Hasselgren PO, Frontera WR, Lee J, Glass DJ, Shoelson SE. IKKbeta/
NF-kappaB activation causes severe muscle wasting in mice. Cell 119:
285–298, 2004.
5. Clarke BA, Drujan D, Willis MS, Murphy LO, Corpina RA, Burova
E, Rakhilin SV, Stitt TN, Patterson C, Latres E, Glass DJ. The E3
Ligase MuRF1 degrades myosin heavy chain protein in dexamethasone-
treated skeletal muscle. Cell Metab 6: 376 –385, 2007.
6. Duclos M, Corcuff JB, Arsac L, Moreau-Gaudry F, Rashedi M, Roger
P, Tabarin A, Manier G. Corticotroph axis sensitivity after exercise in
endurance-trained athletes. Clin Endocrinol (Oxf) 48: 493–501, 1998.
7. Dyer BW, Ferrer FA, Klinedinst DK, Rodriguez R. A noncommercial
dual luciferase enzyme assay system for reporter gene analysis. Anal
Biochem 282: 158 –161, 2000.
8. Furlow JD, Berry DL, Wang Z, Brown DD. A set of novel tadpole
specific genes expressed only in the epidermis are down-regulated by
thyroid hormone during Xenopus laevis metamorphosis. Dev Biol 182:
284 –298, 1997.
9. Furuyama T, Kitayama K, Yamashita H, Mori N. Forkhead transcrip-
tion factor FOXO1 (FKHR)-dependent induction of PDK4 gene expres-
sion in skeletal muscle during energy deprivation. Biochem J 375: 365–
371, 2003.
10. Gan L, Pan H, Unterman TG. Insulin response sequence-dependent and
-independent mechanisms mediate effects of insulin on glucocorticoid-
stimulated insulin-like growth factor binding protein-1 promoter activity.
Endocrinology 146: 4274– 4280, 2005.
11. Glass DJ. Skeletal muscle hypertrophy and atrophy signaling pathways.
Int J Biochem Cell Biol 37: 1974 –1984, 2005.
12. Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL. Atrogin-1,
a muscle-specific F-box protein highly expressed during muscle atrophy.
Proc Natl Acad Sci USA 98: 14440 –14445, 2001.
E795MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
13. Hall RK, Wang XL, George L, Koch SR, Granner DK. Insulin
represses phosphoenolpyruvate carboxykinase gene transcription by caus-
ing the rapid disruption of an active transcription complex: a potential
epigenetic effect. Mol Endocrinol 21: 550 –563, 2007.
14. Hasselgren PO. Glucocorticoids and muscle catabolism. Curr Opin Clin
Nutr Metab Care 2: 201–205, 1999.
15. Hasselgren PO, Fischer JE. Muscle cachexia: current concepts of intra-
cellular mechanisms and molecular regulation. Ann Surg 233: 9 –17, 2001.
16. Hirner S, Krohne C, Schuster A, Hoffmann S, Witt S, Erber R, Sticht
C, Gasch A, Labeit S, Labeit D. MuRF1-dependent regulation of
systemic carbohydrate metabolism as revealed from transgenic mouse
studies. J Mol Biol 379: 666– 677, 2008.
17. Hribal ML, Nakae J, Kitamura T, Shutter JR, Accili D. Regulation of
insulin-like growth factor-dependent myoblast differentiation by Foxo
forkhead transcription factors. J Cell Biol 162: 535–541, 2003.
18. Imai E, Stromstedt PE, Quinn PG, Carlstedt-Duke J, Gustafsson JA,
Granner DK. Characterization of a complex glucocorticoid response unit
in the phosphoenolpyruvate carboxykinase gene. Mol Cell Biol 10: 4712–
4719, 1990.
19. Kamei Y, Miura S, Suzuki M, Kai Y, Mizukami J, Taniguchi T,
Mochida K, Hata T, Matsuda J, Aburatani H, Nishino I, Ezaki O.
Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal
muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes,
and impaired glycemic control. J Biol Chem 279: 41114– 41123, 2004.
20. Kim J, Jia L, Stallcup MR, Coetzee GA. The role of protein kinase A
pathway and cAMP responsive element-binding protein in androgen
receptor-mediated transcription at the prostate-specific antigen locus. J
Mol Endocrinol 34: 107–118, 2005.
21. Kim JJ, Buzzio OL, Li S, Lu Z. Role of FOXO1A in the regulation of
insulin-like growth factor-binding protein-1 in human endometrial cells:
interaction with progesterone receptor. Biol Reprod 73: 833– 839, 2005.
22. Koyama S, Hata S, Witt CC, Ono Y, Lerche S, Ojima K, Chiba T, Doi
N, Kitamura F, Tanaka K, Abe K, Witt SH, Rybin V, Gasch A, Franz
T, Labeit S, Sorimachi H. Muscle RING-finger protein-1 (MuRF1) as a
connector of muscle energy metabolism and protein synthesis. J Mol Biol
376: 1224 –1236, 2008.
23. Krones-Herzig A, Mesaros A, Metzger D, Ziegler A, Lemke U, Brun-
ing JC, Herzig S. Signal-dependent control of gluconeogenic key enzyme
genes through coactivator-associated arginine methyltransferase 1. J Biol
Chem 281: 3025–3029, 2006.
24. Kwon HS, Huang B, Unterman TG, Harris RA. Protein kinase B-alpha
inhibits human pyruvate dehydrogenase kinase-4 gene induction by dexa-
methasone through inactivation of FOXO transcription factors. Diabetes
53: 899 –910, 2004.
25. Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, Price
SR, Mitch WE, Goldberg AL. Multiple types of skeletal muscle atrophy
involve a common program of changes in gene expression. FASEB J 18:
39 –51, 2004.
26. Li P, Lee H, Guo S, Unterman TG, Jenster G, Bai W. AKT-indepen-
dent protection of prostate cancer cells from apoptosis mediated through
complex formation between the androgen receptor and FKHR. Mol Cell
Biol 23: 104 –118, 2003.
27. Lynch GS, Schertzer JD, Ryall JG. Therapeutic approaches for muscle
wasting disorders. Pharmacol Ther 113: 461– 487, 2007.
28. Ma H, Hong H, Huang SM, Irvine RA, Webb P, Kushner PJ, Coetzee
GA, Stallcup MR. Multiple signal input and output domains of the
160-kilodalton nuclear receptor coactivator proteins. Mol Cell Biol 19:
6164 6173, 1999.
29. Martinez E, Wahli W. Cooperative binding of estrogen receptor to
imperfect estrogen-responsive DNA elements correlates with their syner-
gistic hormone-dependent enhancer activity. EMBO J 8: 3781–3791,
1989.
30. Metivier R, Penot G, Hubner MR, Reid G, Brand H, Kos M, Gannon
F. Estrogen receptor-alpha directs ordered, cyclical, and combinatorial
recruitment of cofactors on a natural target promoter. Cell 115: 751–763,
2003.
31. Minetto MA, Lanfranco F, Baldi M, Termine A, Kuipers H, Ghigo E,
Rainoldi A. Corticotroph axis sensitivity after exercise: comparison be-
tween elite athletes and sedentary subjects. J Endocrinol Invest 30:
215–223, 2007.
32. Mitch WE, Goldberg AL. Mechanisms of muscle wasting. The role of
the ubiquitin-proteasome pathway. N Engl J Med 335: 1897–1905,
1996.
33. Nader GA. Concurrent strength and endurance training: from molecules
to man. Med Sci Sports Exerc 38: 1965–1970, 2006.
34. Nasrin N, Ogg S, Cahill CM, Biggs W, Nui S, Dore J, Calvo D, Shi Y,
Ruvkun G, Alexander-Bridges MC. DAF-16 recruits the CREB-binding
protein coactivator complex to the insulin-like growth factor binding
protein 1 promoter in HepG2 cells. Proc Natl Acad Sci USA 97: 10412–
10417, 2000.
35. Nelson JD, Denisenko O, Sova P, Bomsztyk K. Fast chromatin immu-
noprecipitation assay. Nucleic Acids Res 34: e2, 2006.
36. Onuma H, Vander Kooi BT, Boustead JN, Oeser JK, O’Brien RM.
Correlation between FOXO1a (FKHR) and FOXO3a (FKHRL1) binding
and the inhibition of basal glucose-6-phosphatase catalytic subunit gene
transcription by insulin. Mol Endocrinol 20: 2831–2847, 2006.
37. Payvar F, DeFranco D, Firestone GL, Edgar B, Wrange O, Okret S,
Gustafsson JA, Yamamoto KR. Sequence-specific binding of glucocor-
ticoid receptor to MTV DNA at sites within and upstream of the tran-
scribed region. Cell 35: 381–392, 1983.
38. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O,
Bock R, Gass P, Schmid W, Herrlich P, Angel P, Schutz G. DNA
binding of the glucocorticoid receptor is not essential for survival. Cell 93:
531–541, 1998.
39. Reichardt HM, Tuckermann JP, Gottlicher M, Vujic M, Weih F,
Angel P, Herrlich P, Schutz G. Repression of inflammatory responses in
the absence of DNA binding by the glucocorticoid receptor. EMBO J 20:
7168 –7173, 2001.
40. Rena G, Guo S, Cichy SC, Unterman TG, Cohen P. Phosphorylation of
the transcription factor forkhead family member FKHR by protein kinase
B. J Biol Chem 274: 17179 –17183, 1999.
41. Sacheck JM, Ohtsuka A, McLary SC, Goldberg AL. IGF-I stimulates
muscle growth by suppressing protein breakdown and expression of
atrophy-related ubiquitin ligases, atrogin-1 and MuRF1. Am J Physiol
Endocrinol Metab 287: E591–E601, 2004.
42. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh
K, Schiaffino S, Lecker SH, Goldberg AL. Foxo transcription factors
induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal
muscle atrophy. Cell 117: 399– 412, 2004.
43. Schoneveld OJ, Gaemers IC, Lamers WH. Mechanisms of glucocorti-
coid signalling. Biochim Biophys Acta 1680: 114 –128, 2004.
44. Schuur ER, Loktev AV, Sharma M, Sun Z, Roth RA, Weigel RJ.
Ligand-dependent interaction of estrogen receptor-alpha with members
of the forkhead transcription factor family. J Biol Chem 276: 33554–
33560, 2001.
45. So AY, Chaivorapol C, Bolton EC, Li H, Yamamoto KR. Determinants
of cell- and gene-specific transcriptional regulation by the glucocorticoid
receptor. PLoS Genet 3: e94, 2007.
46. So AY, Cooper SB, Feldman BJ, Manuchehri M, Yamamoto KR.
Conservation analysis predicts in vivo occupancy of glucocorticoid recep-
tor-binding sequences at glucocorticoid-induced genes. Proc Natl Acad
Sci USA 105: 5745–5749, 2008.
47. Southgate RJ, Neill B, Prelovsek O, El-Osta A, Kamei Y, Miura S,
Ezaki O, McLoughlin TJ, Zhang W, Unterman TG, Febbraio MA.
FOXO1 regulates the expression of 4E-BP1 and inhibits mTOR sig-
naling in mammalian skeletal muscle. J Biol Chem 282: 21176 –21186,
2007.
48. Stitt TN, Drujan D, Clarke BA, Panaro FJ, Timofeyva Y, Kline
WO, Gonzalez M, Yancopoulos GD, Glass DJ. The IGF-1/PI3K/Akt
pathway prevents expression of muscle atrophy-induced ubiquitin
liganses by inhibiting FOXO transcription factors. Mol Cell 14: 1–14,
2004.
49. Sun L, Trausch-Azar JS, Muglia LJ, Schwartz AL. Glucocorticoids
differentially regulate degradation of MyoD and Id1 by N-terminal ubiq-
uitination to promote muscle protein catabolism. Proc Natl Acad Sci USA
105: 3339 –3344, 2008.
50. Tuckermann JP, Kleiman A, McPherson KG, Reichardt HM. Molec-
ular mechanisms of glucocorticoids in the control of inflammation and
lymphocyte apoptosis. Crit Rev Clin Lab Sci 42: 71–104, 2005.
51. Vander Kooi BT, Onuma H, Oeser JK, Svitek CA, Allen SR,
Vander Kooi CW, Chazin WJ, O’Brien RM. The glucose-6-phos-
phatase catalytic subunit gene promoter contains both positive and
negative glucocorticoid response elements. Mol Endocrinol 19: 3001–
3022, 2005.
52. Vander Kooi BT, Streeper RS, Svitek CA, Oeser JK, Powell DR,
O’Brien RM. The three insulin response sequences in the glucose-6-
E796 MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
phosphatase catalytic subunit gene promoter are functionally distinct.
J Biol Chem 278: 11782–11793, 2003.
53. Wang X, Hu Z, Hu J, Du J, Mitch WE. Insulin resistance accelerates
muscle protein degradation: Activation of the ubiquitin-proteasome pathway
by defects in muscle cell signaling. Endocrinology 147: 4160 4168, 2006.
54. Weinstein SP, Paquin T, Pritsker A, Haber RS. Glucocorticoid-induced
insulin resistance: dexamethasone inhibits the activation of glucose trans-
port in rat skeletal muscle by both insulin- and non-insulin-related stimuli.
Diabetes 44: 441– 445, 1995.
55. Whiteman EL, Cho H, Birnbaum MJ. Role of Akt/protein kinase B in
metabolism. Trends Endocrinol Metab 13: 444 451, 2002.
56. Wright AP, Zilliacus J, McEwan IJ, Dahlman-Wright K, Almlof T,
Carlstedt-Duke J, Gustafsson JA. Structure and function of the glu-
cocorticoid receptor. J Steroid Biochem Mol Biol 47: 11–19, 1993.
57. Yang H, Mammen J, Wei W, Menconi M, Evenson A, Fareed M,
Petkova V, Hasselgren PO. Expression and activity of C/EBPbeta and
delta are upregulated by dexamethasone in skeletal muscle. J Cell Physiol
204: 219 –226, 2005.
58. Yang H, Menconi MJ, Wei W, Petkova V, Hasselgren PO. Dexameth-
asone upregulates the expression of the nuclear cofactor p300 and its
interaction with C/EBPbeta in cultured myotubes. J Cell Biochem 94:
1058 –1067, 2005.
59. Zhao HH, Herrera RE, Coronado-Heinsohn E, Yang MC, Ludes-
Meyers JH, Seybold-Tilson KJ, Nawaz Z, Yee D, Barr FG, Diab SG,
Brown PH, Fuqua SA, Osborne CK. Forkhead homologue in rhabdo-
myosarcoma functions as a bifunctional nuclear receptor-interacting pro-
tein with both coactivator and corepressor functions. J Biol Chem 276:
27907–27912, 2001.
60. Zhao X, Gan L, Pan H, Kan D, Majeski M, Adam SA, Unterman TG.
Multiple elements regulate nuclear/cytoplasmic shuttling of FOXO1: char-
acterization of phosphorylation- and 14-3-3-dependent and -independent
mechanisms. Biochem J 378: 839 849, 2004.
E797MuRF1 GENE REGULATION BY GR AND FOXO1
AJP-Endocrinol Metab VOL 295 OCTOBER 2008 www.ajpendo.org
on October 22, 2008 ajpendo.physiology.orgDownloaded from
... Although hyperalgesia may lead to an increase in the secretion of glucocorticoids, 14 which induce muscle atrophy, 15 we did not observe differences between the pH 4.0 and control groups in terms of plasma corticosterone concentration (t = 2.212; P = 0.0626 vs control, n = 4-5/group) ( Figure 3H). ...
... Therefore, it is possible that the increased proteolysis and reduced protein synthesis in muscles of pH 4.0 group could be mediated by increased sensitivity to glucocorticoids induced by hyperalgesia. This hypothesis was based on the following findings: (a) atrophy was observed only in oxidative muscles after CDH induction, which express more glucocorticoid receptors (GRs) than glycolytic muscles 40 ; (b) there was higher gene expression of Trim63, which has a GR-responsive promoter region, 15 unlike Fbxo32, which remained unchanged in pH 4.0 group muscles; ...
Article
Full-text available
Although it is well established that fibromyalgia (FM) syndrome is characterized by chronic diffuse musculoskeletal hyperalgesia, very little is known about the effect of this pathology on muscle tissue plasticity. Therefore, the present study aimed to characterize the putative alterations in skeletal muscle mass in female rats subjected to a FM model by inducing chronic diffuse hyperalgesia (CDH) through double injections of acidic saline (pH 4.0) into the left gastrocnemius muscle at 5‐day intervals. To determine protein turnover, the total proteolysis, proteolytic system activities and protein synthesis were evaluated in oxidative soleus muscles of pH 7.2 (control) and pH 4.0 groups at 7 days after CDH induction. All animals underwent behavioural analyses of mechanical hyperalgesia, strength and motor performance. Our results demonstrated that, in addition to hyperalgesia, rats injected with acidic saline exhibited skeletal muscle loss, as evidenced by a decrease in the soleus fibre cross‐sectional area. This muscle loss was associated with increased proteasomal proteolysis and expression of the atrophy‐related gene (muscle RING‐finger protein‐1), as well as reduced protein synthesis and decreased protein kinase B/S6 pathway activity. Although the plasma corticosterone concentration did not differ between the control and pH 4.0 groups, the removal of the adrenal glands attenuated hyperalgesia, but it did not prevent the increase in muscle protein loss in acidic saline‐injected animals. The data suggests that the stress‐related hypothalamic–pituitary–adrenal axis is involved in the development of hyperalgesia, but is not responsible for muscle atrophy observed in the FM model induced by intramuscular administration of acidic saline. Although the mechanisms involved in the attenuation of hyperalgesia in rats injected with acidic saline and subjected to adrenalectomy still need to be elucidated, the results found in this study suggest that glucocorticoids may not represent an effective therapeutic approach to alleviate FM symptoms.
... Muscle atrophy F-box (MAFbx) and muscle-specific RING finger protein-1 (MuRF1), two muscle-specific E3 ligases associated with ubiquitin-proteasomal degradation in muscle atrophy, are known as muscle proteolysis markers [15]. In addition, MAFbx and MuRF1 are transcriptionally activated by kruppel-like factor 15 (KLF 15), FoxO1, and FoxO3a against DEX [6,16,17]. Moreover, an increased expression of regulated in development and DNA damage responses 1 (REDD1) (also known as DNA damage-inducible transcript 4 (DDIT4)) and KLF15 inhibits mTORC1 activity [18,19]. ...
... MAFbx and MuRF1 are transcriptionally activated by KLF 15, FoxO1, and FoxO3a in response to DEX [6,16,17]. We also observed that ECR repressed the mRNA expression of KLF15 and REDD1 in DEX-induced muscle atrophy. ...
Article
Full-text available
Sarcopenia is a progressive muscle disease characterized by the loss of skeletal muscle mass, strength, function, and physical performance. Since the disease code was assigned, attention has been focused on natural products that can protect against muscle atrophy. Cibotium barometz (Cibotium Rhizome) has been used as an herbal medicine for the treatment of bone or joint diseases in Asian countries. However, no studies have identified the mechanism of action of Cibotium Rhizome on muscle atrophy related to sarcopenia at the site of myotubes. The aim of this study was to investigate the improvement effect of the ethanol extract of Cibotium Rhizome (ECR) on dexamethasone-induced muscle atrophy in an in vitro cell model, i.e., the C2C12 myotubes. High-performance liquid chromatography was performed to examine the phytochemicals in ECR. Seven peaks in the ECR were identified, corresponding to the following compounds: protocatechuic acid, (+)-catechin hydrate, p-coumaric acid, ellagic acid, chlorogenic acid, caffeic acid, and ferulic acid. In atrophy-like conditions induced by 100 μM dexamethasone for 24 h in C2C12, ECR increased the expression of the myosin heavy chain, p-Akt, the p-mammalian target of rapamycin (mTOR), p-p70S6K, and repressed the expression of regulated in development and DNA damage responses 1 (REDD1), kruppel-like factor 15 (KLF 15), muscle atrophy F-box, and muscle-specific RING finger protein-1 in C2C12. In addition, ECR alleviated dexamethasone-induced muscle atrophy by repressing REDD1 and KLF15 transcription in C2C12 myotubes, indicating the need for further studies to provide a scientific basis for the development of useful therapeutic agents using ECR to alleviate the effects of skeletal muscle atrophy or sarcopenia.
... In recent years, bile acids have been found to affect glucose metabolism, insulin sensitivity, metabolic dysfunction, mass, and atrophy of skeletal muscle [32,[37][38][39]. Meanwhile, regarding branched-chain amino acids, it has been reported that skeletal muscle growth and development are closely related to branched-chain amino acid metabolism [40,41]. Therefore, an intriguing avenue for future exploration is whether metabolites other than acetate produced by gut microbiota also play a role in regulating skeletal muscle growth and development. ...
Article
Full-text available
The normal growth and development of skeletal muscle is essential for the health of the body. The regulation of skeletal muscle by intestinal microorganisms and their metabolites has been continuously demonstrated. Acetate is the predominant short-chain fatty acids synthesized by gut microbiota through the fermentation of dietary fiber; however, the underlying molecular mechanisms governing the interaction between acetate and skeletal muscle during the rapid growth stage remains to be further elucidated. Herein, specific pathogen-free (SPF) mice, germ-free (GF) mice, and germ-free mice supplemented with sodium acetate (GS) were used to evaluate the effects of acetate on the skeletal muscle growth and development of young mice with gut microbiota deficiency. We found that the concentration of serum acetate, body mass gain, succinate dehydrogenase activity, and expression of the myogenesis maker gene of skeletal muscle in the GS group were higher than those in the GF group, following sodium acetate supplementation. Furthermore, the transcriptome analysis revealed that acetate activated the biological processes that regulate skeletal muscle growth and development in the GF group, which are otherwise inhibited due to a gut microbiota deficiency. The in vitro experiment showed that acetate up-regulated Gm16062 to promote skeletal muscle cell differentiation. Overall, our findings proved that acetate promotes skeletal muscle growth and development in young mice via increasing Gm16062 expression.
... In recent years, bile acids were demonstrated affect glucose metabolism, insulin sensitivity, energy metabolism, mass and atrophy of skeletal muscle [32,[36][37][38]. And branched chain amino acids, it was reported that skeletal muscle growth and development are closely related to branched chain amino acid metabolism [39,40]. Therefore, an intriguing avenue for future exploration is whether metabolites other than acetate produced by the gut microbiota also play a role in regulating skeletal muscle growth and development. ...
Preprint
Full-text available
The normal growth and development of skeletal muscle is essential for the health of the body. The regulation of skeletal muscle by intestinal microorganisms and their metabolites has been continuously revealed. Acetate is the predominant short-chain fatty acid synthesized by gut microbiota through the fermentation of dietary fiber, however, the underlying molecular mechanisms governing the interaction between acetate and skeletal muscle during rapid growth stage remain to be further elucidated; Here, specific pathogen-free (SPF) mice, germ-free (GF) mice, and germ-free mice supplemented with sodium acetate (GS) were used to evaluate the effects of acetate on skeletal muscle growth and development of young mice with gut microbiota deficiency; We found the concentration of serum acetate, body mass gain, succinate dehydrogenase activity and expression of myogenesis maker gene of skeletal muscle in the GS group was higher than GF group following sodium acetate supplement. Furthermore, the transcriptome analysis revealed that acetate activated biological processes regulating skeletal muscle growth and development in GF group that are inhibited due to gut microbiota deficiency. In vitro experiment, we proved that acetate up-regulated Gm16062 to promote skeletal muscle cell differentiation; Overall, our findings proved that acetate promote skeletal muscle growth and development in young mice via increase Gm16062 expression.
... SiRNA silencing of FoxO1 rescued the AGE effect on muscle atrophy signifying that AGE production in T2D may promote muscle wasting via FoxO1. Additionally, FoxO1 has been shown to decrease muscle mass by upregulating the ubiquitin ligase MuRF1 and the liposomal protease cathepsin L (96,97). Skeletal muscle specific knockout studies further support a role for FoxOs in mediating muscle atrophy. ...
Article
Full-text available
Forkhead box O (FoxO) proteins are transcription factors that mediate many aspects of physiology and thus have been targeted as therapeutics for several diseases including metabolic disorders such as type 2 diabetes mellitus (T2D). The role of FoxO1 in metabolism has been well studied, but recently FoxO1’s potential for diabetes prevention and therapy has been debated. For example, studies have shown that increased FoxO1 activity in certain tissue types contributes to T2D pathology, symptoms, and comorbidities, yet in other tissue types elevated FoxO1 has been reported to alleviate symptoms associated with diabetes. Furthermore, studies have reported opposite effects of active FoxO1 in the same tissue type. For example, in the liver, FoxO1 contributes to T2D by increasing hepatic glucose production. However, FoxO1 has been shown to either increase or decrease hepatic lipogenesis as well as adipogenesis in white adipose tissue. In skeletal muscle, FoxO1 reduces glucose uptake and oxidation, promotes lipid uptake and oxidation, and increases muscle atrophy. While many studies show that FoxO1 lowers pancreatic insulin production and secretion, others show the opposite, especially in response to oxidative stress and inflammation. Elevated FoxO1 in the hypothalamus increases the risk of developing T2D. However, increased FoxO1 may mitigate Alzheimer’s disease, a neurodegenerative disease strongly associated with T2D. Conversely, accumulating evidence implicates increased FoxO1 with Parkinson’s disease pathogenesis. Here we review FoxO1’s actions in T2D conditions in metabolic tissues that abundantly express FoxO1 and highlight some of the current studies targeting FoxO1 for T2D treatment.
Article
Full-text available
Synthetic glucocorticoids (GC), such as dexamethasone, are extensively used to treat chronic inflammation and autoimmune disorders. However, long-term treatments are limited by various side effects, including muscle atrophy. GC activities are mediated by the glucocorticoid receptor (GR), that regulates target gene expression in various tissues in association with cell-specific co-regulators. Here we show that GR and the lysine-specific demethylase 1 (LSD1) interact in myofibers of male mice, and that LSD1 connects GR-bound enhancers with NRF1-associated promoters to stimulate target gene expression. In addition, we unravel that LSD1 demethylase activity is required for triggering starvation- and dexamethasone-induced skeletal muscle proteolysis in collaboration with GR. Importantly, inhibition of LSD1 circumvents muscle wasting induced by pharmacological levels of dexamethasone, without affecting their anti-inflammatory activities. Thus, our findings provide mechanistic insights into the muscle-specific GC activities, and highlight the therapeutic potential of targeting GR co-regulators to limit corticotherapy-induced side effects.
Article
Fibroblast growth factor-21 (FGF21) is an intercellular signaling molecule secreted by metabolic organs, including skeletal muscle, in response to intracellular stress. FGF21 crosses the blood brain barrier and acts via the nervous system to coordinate aspects of the adaptive starvation response, including increased lipolysis, gluconeogenesis, fatty acid oxidation, and activation of the hypothalamic-pituitary-adrenocortical (HPA) axis. Given its beneficial effects for hepatic lipid metabolism, pharmaceutical FGF21 analogues are in clinical trials treatment of fatty liver disease. We predicted pharmacologic treatment with FGF21 increases HPA axis activity and skeletal muscle glucocorticoid signaling and induces skeletal muscle atrophy in mice. Here we found a short course of systemic FGF21 treatment decreased muscle protein synthesis and reduced tibialis anterior weight; this was driven primarily by its effect in female mice. Similarly, intracerebroventricular FGF21 reduced TA muscle fiber cross sectional area; this was more apparent among female mice compared to male littermates. In agreement with the reduced muscle mass, the topmost enriched metabolic pathways in plasma collected from FGF21-treated females were related to amino acid metabolism, and the relative abundance of plasma proteinogenic amino acids were increased up to three-fold. FGF21 treatment increased hypothalamic Crh mRNA, plasma corticosterone, and adrenal weight, and increased expression of glucocorticoid receptor target genes known to reduce muscle protein synthesis and/or promote degradation. Given the proposed use of FGF21 analogues for the treatment of metabolic disease, the study is both physiologically relevant and may have important clinical implications.
Article
Full-text available
Cancer cachexia is a multi-organ syndrome and closely related to changes in signal communication between organs, which is mediated by cancer cachexia factors. Cancer cachexia factors, being the general name of inflammatory factors, circulating proteins, metabolites, and microRNA secreted by tumor or host cells, play a role in secretory or other organs and mediate complex signal communication between organs during cancer cachexia. Cancer cachexia factors are also a potential target for the diagnosis and treatment. The pathogenesis of cachexia is unclear and no clear effective treatment is available. Thus, the treatment of cancer cachexia from the perspective of the tumor ecosystem rather than from the perspective of a single molecule and a single organ is urgently needed. From the point of signal communication between organs mediated by cancer cachexia factors, finding a deeper understanding of the pathogenesis, diagnosis, and treatment of cancer cachexia is of great significance to improve the level of diagnosis and treatment. This review begins with cancer cachexia factors released during the interaction between tumor and host cells, and provides a comprehensive summary of the pathogenesis, diagnosis, and treatment for cancer cachexia, along with a particular sight on multi-organ signal communication mediated by cancer cachexia factors. This summary aims to deepen medical community's understanding of cancer cachexia and may conduce to the discovery of new diagnostic and therapeutic targets for cancer cachexia.
Article
Full-text available
The musculoskeletal system is important for balancing metabolic activity and maintaining health. Recent studies have shown that distortions in homeostasis of the intestinal microbiota are correlated with or may even contribute to abnormalities in musculoskeletal system function. Research has also shown that the intestinal flora and its secondary metabolites can impact the musculoskeletal system by regulating various phenomena, such as inflammation and immune and metabolic activities. Most of the existing literature supports that reasonable nutritional intervention helps to improve and maintain the homeostasis of intestinal microbiota, and may have a positive impact on musculoskeletal health. The purpose of organizing, summarizing and discussing the existing literature is to explore whether the intervention methods, including nutritional supplement and moderate exercise, can affect the muscle and bone health by regulating the microecology of the intestinal flora. More in-depth efficacy verification experiments will be helpful for clinical applications.
Article
Full-text available
In a search for novel transcriptional intermediary factors for the estrogen receptor (ER), we used the ligand-binding domain and hinge region of ER as bait in a yeast two-hybrid screen of a cDNA library derived from tamoxifen-resistant MCF-7 human breast tumors from an in vivo athymic nude mouse model. Here we report the isolation and characterization of the forkhead homologue in rhabdomyosarcoma (FKHR), a recently described member of the hepatocyte nuclear factor 3/forkhead homeotic gene family, as a nuclear hormone receptor (NR) intermediary protein. FKHR interacts with both steroid and nonsteroid NRs, although the effect of ligand on this interaction varies by receptor type. The interaction of FKHR with ER is enhanced by estrogen, whereas its interaction with thyroid hormone receptor and retinoic acid receptor is ligand-independent. In addition, FKHR differentially regulates the transactivation mediated by different NRs. Transient transfection of FKHR into mammalian cells dramatically represses transcription mediated by the ER, glucocorticoid receptor, and progesterone receptor. In contrast, FKHR stimulates rather than represses retinoic acid receptor- and thyroid hormone receptor-mediated transactivation. Most intriguingly, overexpression of FKHR dramatically inhibits the proliferation of ER-dependent MCF-7 breast cancer cells. Therefore, FKHR represents a bifunctional NR intermediary protein that can act as either a coactivator or corepressor, depending on the receptor type.
Article
Full-text available
The mammalian target of rapamycin (mTOR) is regulated by growth factors to promote protein synthesis. In mammalian skeletal muscle, the Forkhead-O1 transcription factor (FOXO1) promotes catabolism by activating ubiquitin-protein ligases. Using C2C12 mouse myoblasts that stably express inducible FOXO1-ER fusion proteins and transgenic mice that specifically overexpress constitutively active FOXO1 in skeletal muscle (FOXO(++/+)), we show that FOXO1 inhibits mTOR signaling and protein synthesis. Activation of constitutively active FOXO1 induced the expression of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) mRNA via binding to the promoter. This resulted in an increased total 4E-BP1 abundance and a reduced 4E-BP1 (Thr-37/46) phosphorylation. The reduction in 4E-BP1 phosphorylation was associated with a reduction in the abundance of Raptor and mTOR proteins, Raptor-associated mTOR, reduced phosphorylation of the downstream protein p70S6 kinase, and attenuated incorporation of [(14)C]phenylalanine into protein. The FOXO(++/+) mice, characterized by severe skeletal muscle atrophy, displayed similar patterns of mRNA expression and protein abundance to those observed in the constitutively active FOXO1 C2C12 myotubes. These data suggest that FOXO1 may be an important therapeutic target for human diseases where anabolism is impaired.
Article
Full-text available
The minimal DNA sequence required for glucocorticoid induction of the phosphoenolpyruvate carboxykinase (PEPCK) gene in H4IIE rat hepatoma cells was defined. This novel glucocorticoid response unit (GRU) spans about 110 base pairs (bp) and includes two receptor-binding elements plus two accessory factor-binding elements. Purified glucocorticoid receptor bound to two regions (GR1 and GR2) between -395 and -349 bp relative to the transcription start site. Factors in crude rat liver nuclear extract bound to DNA in the regions -455 to -431 and -420 to -403 bp, which are designated accessory factor 1 (AF1) and accessory factor 2 (AF2) elements, respectively. Gel retardation analysis revealed that at least two proteins bound to AF1 and that they were distinct from the protein(s) that bound to AF2. Various combinations of GR1, GR2, AF1, and AF2 were fused to the chloramphenicol acetyltransferase (CAT) reporter gene and cotransfected with a glucocorticoid receptor expression plasmid (pSVGR1) into H4IIE cells to identify the functional GRU. Neither the glucocorticoid receptor binding region nor the accessory factor binding region alone was sufficient to confer glucocorticoid responsiveness. The two components of the glucocorticoid receptor binding region functioned independently, and each accounted for half of the maximal response, provided the accessory factor elements were present. Similarly, deletion of either AF1 or AF2 diminished glucocorticoid induction of the PEPCK gene to approximately half of the maximum. We propose that the complex PEPCK gene GRU provides the stringent regulation required of this critical enzyme in liver.
Article
Full-text available
The Xenopus vitellogenin (vit) gene B1 estrogen-inducible enhancer is formed by two closely adjacent 13 bp imperfect palindromic estrogen-responsive elements (EREs), i.e. ERE-2 and ERE-1, having one and two base substitutions respectively, when compared to the perfect palindromic consensus ERE (GGTCANNNTGACC). Gene transfer experiments indicate that these degenerated elements, on their own, have a low or no regulatory capacity at all, but in vivo act together synergistically to confer high receptor- and hormone-dependent transcription activation to the heterologous HSV thymidine kinase promoter. Thus, the DNA region upstream of the vitB1 gene comprising these two imperfect EREs separated by 7 bp, was called the vitB1 estrogen-responsive unit (vitB1 ERU). Using in vitro protein-DNA interaction techniques, we demonstrate that estrogen receptor dimers bind cooperatively to the imperfect EREs of the vitB1 ERU. Binding of a first receptor dimer to the more conserved ERE-2 increases approximately 4- to 8-fold the binding affinity of the receptor to the adjacent less conserved ERE-1. Thus, we suggest that the observed synergistic estrogen-dependent transcription activation conferred by the pair of hormone-responsive DNA elements of the vit B1 ERU is the result of cooperative binding of two estrogen receptor dimers to these two adjacent imperfect EREs.
Article
Transcriptional regulation by the glucocorticoid receptor (GR) is essential for survival. Since the GR can influence transcription both through DNA-binding-dependent and -independent mechanisms, we attempted to assess their relative importance in vivo. In order to separate these modes of action, we introduced the point mutation A458T into the GR by gene targeting using the Cre/loxP system. This mutation impairs dimerization and therefore GRE-dependent transactivation while functions that require cross-talk with other transcription factors, such as transrepression of AP-1-driven genes, remain intact. In contrast to GR-/- mice, these mutants termed GRdim are viable, revealing the in vivo relevance of DNA-binding-independent activities of the GR.
Article
Gene regulation by steroid hormones is mediated through an interaction of the hormone receptors with DNA regulatory sequences called hormone regulatory or responsive elements (HRE). An analysis of the HRE's in the DNA of mouse mammary tumour provirus, human metallothionein IIA gene, chicken lysozyme gene, chicken and Xenopus vitellogenin genes, growth hormones genes, Moloney murine sarcoma provirus, rabbit uteroglobin gene, rat tyrosine aminotransferase gene, rat tryptophan oxygenase gene and rat acidic glycoprotein gene, yields the following consensus for positively modulated glucocorticoid responsive elements (GRE): 5'-GGTACAnnnTGTTCT-3'. This element can also mediate induction by progesterone and probably by androgens, but not by estrogens. Detailed analysis of the DNA protection pattern suggests that a dimer of the hormone receptor interacts with this palindromic 15-mer. In genes that are negatively regulated by glucocorticoids an imperfect copy of the GRE is found, and repression is probably due to competition between hormone receptor and other transcription factors or enhancer binding proteins for binding to overlapping DNA sequences. The receptors without bound hormone are able to interact specifically with DNA in vitro, but binding of hormone is needed for transcriptional activation in vivo. This could be due, at least in part, to changes in the rate parameters of the receptor-DNA interaction induced by binding of the hormone to the receptor. The possible role of precise chromatin organization in glucocorticoid induction is discussed on the basis of the nucleosome phasing found in the LTR region of mouse mammary tumour virus.
Article
The glucocorticoid receptor protein is a transcriptional regulatory protein that interacts with specific enhancer sequences. A stoichiometric analysis of the interaction indicates an equimolar relationship between the receptor protein and the specific enhancer sequences. The activity of the receptor protein is itself regulated by the binding of glucocorticoids. The two functional domains (DNA-binding and steroid-binding) are adjacent and lie within the C-terminal half of the receptor protein. The N-terminal half of the protein appears to modulate the interaction with DNA but does not have any role in the binding of the steroid. The functional domains have also been defined at the sequence level.
Article
Glucocorticoid receptor protein stimulates transcription initiation within murine mammary tumor virus (MTV) DNA sequences in vivo, and interacts selectively with MTV DNA in vitro. We mapped and compared five regions of MTV DNA that are bound specifically by purified receptor; one resides upstream of the transcription start site, and the others are distributed within transcribed sequences between 4 and 8 kb from the initiation site. Each region contains at least two strong binding sites for receptor, which itself appears to be a tetramer of 94,000 dalton hormone-binding subunits. Three of the five binding regions contain nine nuclease footprints that lack extensive homology, although a family of related octanucleotides can be discerned. Receptor interacts with the different regions with similar efficiencies, suggesting that receptor affinity for upstream and internal regions may differ by less than one order of magnitude. Moreover, each region appears to be bound independent of the others. A restriction fragment containing four footprint sequences from one of the regions has previously been shown to act in vivo as a receptor-dependent transcriptional enhancer element, implying that the binding sites detected in vitro may be biologically functional.