ArticlePDF Available

Expression and Regulation of Corticotropin-Releasing Factor Receptor Type 2β in Developing and Mature Mouse Skeletal Muscle

Authors:

Abstract and Figures

Corticotropin-releasing factor receptor type 2 (CRFR2) is highly expressed in skeletal muscle (SM) tissue where it is suggested to inhibit interactions between insulin signaling pathway components affecting whole-body glucose homeostasis. However, little is known about factors regulating SM CRFR2 expression. Here, we demonstrate the exclusive expression of CRFR2, and not CRFR1, in mature SM tissue using RT-PCR and ribonuclease protection assays and report a differential expression of CRF receptors during C2C12 myogenic differentiation. Whereas C2C12 myoblasts exclusively express CRFR1, the C2C12 myotubes solely express CRFR2. Using cAMP luciferase assays and calcium mobilization measurements, we further demonstrate the functionality of these differentially expressed receptors. Using luciferase reporter assays we show a differential activation of CRFR promoters during myogenic differentiation. Transfections with different fragments of the 5'-flanking region of the mCRFR2β gene fused to a luciferase reporter gene show a promoter-dependent expression of the reporter gene and reveal the importance of the myocyte enhancer factor 2 consensus sequence located at the 3'-proximal region of CRFR2β promoter. Furthermore, we demonstrate that CRFR2 gene transcription in the mature mouse is stimulated by both high-fat diet and chronic variable stress conditions. Performing a whole-genome expression microarray analysis of SM tissues obtained from CRFR2-null mice or wild-type littermates revealed a robust reduction in retinol-binding protein 4 expression levels, an adipokine whose serum levels are elevated in insulin-resistant states. In correlation with the SM CRFR2β levels, the SM retinol-binding protein 4 levels were also elevated in mice subjected to high-fat diet and chronic variable stress conditions. The current findings further position the SM CRFR2 pathways as a relevant physiological system that may affect the known reciprocal relationship between psychological and physiological challenges and the metabolic syndrome.
Expression of mCRFR2 mRNA in mouse SM and differential expression of mCRFR1 and mCRFR2 mRNA during C2C12 cells myogenic differentiation. A, Representative image of electrophoretic analysis of the semiquantitative RT-PCR products of mCRFR2 ( upper panel ), mCRFR1 ( middle panel ), and the ribosomal protein S16 ( lower panel ) in the mouse SM. Brain samples served as positive controls for both CRFR1 and CRFR2 gene expression. PCR without reverse transcriptase (RT) enzyme ( Ϫ R.T) or without cDNA ( Ϫ cDNA) served as negative controls. B, Representative image of RNase protection assay of mCRFR1 ( right panel ) and mCRFR2 ( left panel) mRNA. SM total RNA was hybridized with the mCRFR1 ( right panel ), mCRFR2 ( left panel ), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) ( both panels ) antisense probes. Brain and pituitary gland served as positive controls for CRFR2 and CRFR1 gene expression, respectively. C, Representative image of electrophoretic analysis of the semiquantitative RT-PCR products of mCRFR1, mCRFR2, muscle differentiation markers, MyoD, myogenin, and Id2 and the ribosomal protein S16 in C2C12 myoblasts cultured in differentiation media (DM). RNA extracted from C2C12 myoblasts cultured in DM (containing 2% horse serum) for 0 – 6 d were reverse transcribed to generate cDNA, which were used as templates to the PCR using specific primers for mCRFR1, mCRFR2, MyoD, myogenin, Id2, and the ribosomal protein S16 that served as an internal control. RNA extracted from mouse SM and brain served as positive controls. PCR without RT enzyme ( Ϫ R.T) or without cDNA ( Ϫ cDNA) served as negative controls. D, The bands were quantified, and the normalized values (relative to the control S16 expression) are presented as fold increase. Three independent experiments were conducted and showed similar kinetic of gene expression.
… 
Content may be subject to copyright.
Expression and Regulation of Corticotropin-Releasing
Factor Receptor Type 2
in Developing and Mature
Mouse Skeletal Muscle
Yael Kuperman, Orna Issler, Joan Vaughan, Louise Bilezikjian, Wylie Vale,
and Alon Chen
Department of Neurobiology (Y.K., O.I., A.C.), Weizmann Institute of Science, Rehovot, 76100, Israel;
and Clayton Foundation Laboratories for Peptide Biology (J.V., L.B., W.V.), Salk Institute for Biological
Studies, La Jolla, California
Corticotropin-releasing factor receptor type 2 (CRFR2) is highly expressed in skeletal muscle (SM)
tissue where it is suggested to inhibit interactions between insulin signaling pathway components
affecting whole-body glucose homeostasis. However, little is known about factors regulating SM
CRFR2 expression. Here, we demonstrate the exclusive expression of CRFR2, and not CRFR1, in
mature SM tissue using RT-PCR and ribonuclease protection assays and report a differential
expression of CRF receptors during C2C12 myogenic differentiation. Whereas C2C12 myoblasts
exclusively express CRFR1, the C2C12 myotubes solely express CRFR2. Using cAMP luciferase assays
and calcium mobilization measurements, we further demonstrate the functionality of these dif-
ferentially expressed receptors. Using luciferase reporter assays we show a differential activation
of CRFR promoters during myogenic differentiation. Transfections with different fragments of the
5-flanking region of the mCRFR2
gene fused to a luciferase reporter gene show a promoter-
dependent expression of the reporter gene and reveal the importance of the myocyte enhancer
factor 2 consensus sequence located at the 3-proximal region of CRFR2
promoter. Furthermore,
we demonstrate that CRFR2 gene transcription in the mature mouse is stimulated by both high-fat
diet and chronic variable stress conditions. Performing a whole-genome expression microarray
analysis of SM tissues obtained from CRFR2-null mice or wild-type littermates revealed a robust
reduction in retinol-binding protein 4 expression levels, an adipokine whose serum levels are
elevated in insulin-resistant states. In correlation with the SM CRFR2
levels, the SM retinol-
binding protein 4 levels were also elevated in mice subjected to high-fat diet and chronic variable
stress conditions. The current findings further position the SM CRFR2 pathways as a relevant
physiological system that may affect the known reciprocal relationship between psychological
and physiological challenges and the metabolic syndrome. (Molecular Endocrinology 25: 157–169,
2011)
Abdominal obesity and insulin resistance have each been
proposed as the primary factors underlying metabolic
syndrome (1, 2). Skeletal muscle (SM) comprises the largest
insulin-sensitive tissue in humans, and thus, insulin resis-
tance in this organ impacts whole-body glucose homeostasis
(3). Insulin resistance in SM was proposed to promote
atherogenic dyslipidemia by decreasing muscle glycogen
synthesis and elevating hepatic de novo lipid synthesis and
very-low-density lipoprotein production (2).
The corticotropin-releasing factor (CRF)/urocortin
(Ucn) family of peptides and receptors is involved in the
maintenance and adaptive responses necessary for energy
homeostasis (4–11). The CRF/Ucn family of neuropep-
tides signals through the activation of two G protein-
ISSN Print 0888-8809 ISSN Online 1944-9917
Printed in U.S.A.
Copyright © 2011 by The Endocrine Society
doi: 10.1210/me.2010-0308 Received August 2, 2010. Accepted October 14, 2010.
First Published Online November 17, 2010
Abbreviations: Ant, Antalarmin; Ast 2b, Astressin 2B; CHO, Chinese hamster ovary; CRE,
cAMP-responsive element; CRF, corticotropin-releasing factor; CRFR2, corticotropin-re-
leasing factor receptor type 2; CVS, chronic variable stress; DM, differentiation medium;
HFD, high-fat diet; HPRT1, hypoxanthine guanine phosphoribosyl transferase 1; iv, inser-
tion variant; KO, knockout; MEF, myocyte enhancer factor; RBP4, retinol-binding protein
4; RNase, ribonuclease; SM, skeletal muscle; Ucn, urocortin; WT, wild type.
ORIGINAL RESEARCH
Mol Endocrinol, January 2011, 25(1):157–169 mend.endojournals.org 157
coupled receptors, CRF receptor type 1 (CRFR1) (12–14)
and CRF receptor type 2, CRFR2 (15–18). Mouse
CRFR2 has three apparent splice variants, which results
in two putative receptor proteins of 411 and 431 amino
acids (CRFR2
and CRFR2
, respectively) and in a 422-
amino acid insertion-variant (iv) with dominant-negative
activity. In rodents, CRFR2
is predominantly expressed
in the brain (19). The CRFR2
splice variant is expressed
primarily in the SM, the heart, the brain choroid plexus, the
gastrointestinal tract, and the skin (17, 20, 21) whereas
ivCRFR2
is exclusively expressed in the heart (22).
In SM tissue, CRFR2
was suggested to be involved in
different cellular processes. SM CRFR2
activation was
suggested to impede glucose metabolism. CRFR2-null
mice have enhanced glucose tolerance, increased insulin
sensitivity and are protected from high-fat diet-induced
insulin resistance (6). Ucn2, which is highly expressed in
SM tissue (23) and most likely serves as the endogenous
ligand for SM CRFR2
, inhibits the interactions between
insulin-signaling pathway components and insulin-in-
duced glucose uptake in cultured SM cells, and in C2C12
myotubes (8). The Ucn2-null mice exhibit increased insu-
lin sensitivity and are protected from fat-induced insulin
resistance (8). In addition, CRFR2
activation was dem-
onstrated to increase SM mass (24), reduce SM mass loss
in atrophying SM due to denervation or casting, and to
increase nonatrophying SM mass (25).
Given the importance of CRFR2 in regulating the cen-
tral stress response and its beneficial effect on cardiovas-
cular function (26), the regulation of its hypothalamic
and heart expression has been extensively studied (Refs.
27–31 and Refs. 22 and 32–35, respectively). However,
little is known regarding factors regulating SM CRFR2
expression. Here, we demonstrate the differential expres-
sion of CRFR1 and CRFR2 mRNA during C2C12 myo-
genic differentiation. The functional signaling of those
receptors was determined, and promoter analysis studies
demonstrated the importance of muscle-specific tran-
scription factors putative binding sites. Additionally, we
show the in vivo regulation of SM CRFR2
mRNA by
chronic physiological or psychological stressors and its
association with insulin-resistant states.
Results
Differential expression of CRFR1 and CRFR2 during
myogenic differentiation
To verify expression of SM CRFRs, total RNA pre-
pared from SM and brain tissues was reverse transcribed
to generate cDNAs. The cDNA products were used as
templates for specific semiquantitative RT-PCR demon-
strating selective CRFR2 expression in SM tissue whereas
the brain cDNA served as a positive control for CRFR1
and CRFR2 expression (Fig. 1A). The selective expression
of CRFR2, and not CRFR1, in SM tissue was further
verified using ribonuclease (RNase) protection assay (Fig.
1B). The multinucleated SM fibers are formed in succes-
sive distinct steps involving different types of myoblasts
(36). For in vitro investigation of the molecular basis of
SM cell differentiation, C2C12 cells, mouse-derived myo-
blasts that can be propagated as undifferentiated mono-
nuclear cells in serum, serve as a useful experimental
model. On serum withdrawal, muscle-specific genes are
expressed leading to the formation of differentiated
multinucleated myotubes (37, 38). To study the CRFR2
expression profile during myogenic differentiation, RNA
extracted from C2C12 myoblasts at different time points
during myogenic differentiation was reverse transcribed
and used as a template for semiquantitative RT-PCR. The
myogenic determination factors MyoD and myogenin, as
well as the negative regulator of myogenesis, Id2 (39),
were used to monitor the differentiation process. Unex-
pectedly, C2C12 myoblasts were found to exclusively ex-
press CRFR1 whereas C2C12 myotubes were found to
exclusively express CRFR2 (Fig. 1C). The time-depen-
dent differential expression of the two-receptor forms can
be observed during the differentiation process (Fig. 1, C and
D). CRFR2 shows expression kinetics similar to MyoD and
myogenin expression profiles whereas CRFR1 expression
mirrors the expression profile Id2 (Fig. 1, C and D).
C2C12 cells were further used for demonstrating
CRFR1- and CRFR2-selective activation in nondifferenti-
ated (myoblasts) or differentiated (myotubes) state. Recep-
tor functionality was demonstrated by measuring the acti-
vation of the cAMP and calcium pathways using CRF or
Ucn2/3, which are specific ligands for CRFR1 and CRFR2,
respectively (4042). Nondifferentiated C2C12 cells were
transfected with a luciferase reporter containing a fragment
of the EVX1 gene that contains a cAMP-responsive element
(CRE) site. Luciferase activity was used as a measure of
receptor activation and was determined after4hoftreat-
ment with vehicle, and various doses of CRF, or Ucn3 in
nondifferentiated, or 48-h differentiated, C2C12 cells. The
CRE-luciferase reporter gene was differentially activated in
the myoblasts and myotubes after stimulation with CRF or
Ucn3, respectively (Fig. 2A). In myoblasts, CRF signaling
induced CRE-luciferase activity, which was blocked by the
CRFR1-specific antagonist Antalarmin (Ant), whereas in
the myotubes, Ucn3 signaling induced CRE-luciferase activ-
ity that was blocked by the CRFR2-specific antagonist As-
tressin 2B (Ast 2B).
Additionally, calcium mobilization kinetics were mea-
sured in nondifferentiated (myoblasts) or differentiated
(myotubes) C2C12 cells by CRF or Ucn2 (Fig. 2B). Acti-
158 Kuperman et al. Expression and Regulation of Muscle CRFR2
Mol Endocrinol, January 2011, 25(1):157–169
vation of calcium flux was differentially activated in the
myoblasts and myotubes after stimulation with CRF.
CRF strongly activated calcium flux in myoblasts, but not
in myotubes, (Fig. 2B, a and b). Ucn2 activated calcium
flux to a lesser extent in myoblasts, probably due to its
low affinity for CRFR1, but did not activate calcium flux
in myotubes (Fig. 2B, c and d). To further explore this
phenomenon, the calcium mobilization kinetic studies
were duplicated in Chinese hamster ovary (CHO) cells
stably expressing CRFR1 or CRFR2 treated with CRF or
Ucn2, respectively (Fig. 2C). Only CRF activation of
CHO cells expressing CRFR1, but not Ucn2 activation of
CHO cells expressing CRFR2, promoted calcium mobi-
lization. Demonstrating that CRFR1, but not CRFR2,
activation will promote calcium mobilization supports
the finding of differential expression of the CRFRs during
myogenic differentiation.
Differential activation of mCRFR1 and mCRFR2
promoters during C2C12 myoblast differentiation
To explore the molecular mechanisms mediating the
differential regulation of CRFR1 and CRFR2 during
myogenic differentiation, and to examine whether the dif-
ferential expression is regulated at the promoter level, we
FIG. 1. Expression of mCRFR2 mRNA in mouse SM and differential expression of mCRFR1 and mCRFR2 mRNA during C2C12 cells myogenic
differentiation. A, Representative image of electrophoretic analysis of the semiquantitative RT-PCR products of mCRFR2 (upper panel), mCRFR1
(middle panel), and the ribosomal protein S16 (lower panel) in the mouse SM. Brain samples served as positive controls for both CRFR1 and CRFR2
gene expression. PCR without reverse transcriptase (RT) enzyme (R.T) or without cDNA (cDNA) served as negative controls. B, Representative
image of RNase protection assay of mCRFR1 (right panel) and mCRFR2 (left panel) mRNA. SM total RNA was hybridized with the mCRFR1 (right
panel), mCRFR2 (left panel), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (both panels) antisense probes. Brain and pituitary gland
served as positive controls for CRFR2 and CRFR1 gene expression, respectively. C, Representative image of electrophoretic analysis of the
semiquantitative RT-PCR products of mCRFR1, mCRFR2, muscle differentiation markers, MyoD, myogenin, and Id2 and the ribosomal protein S16
in C2C12 myoblasts cultured in differentiation media (DM). RNA extracted from C2C12 myoblasts cultured in DM (containing 2% horse serum) for
0–6 d were reverse transcribed to generate cDNA, which were used as templates to the PCR using specific primers for mCRFR1, mCRFR2, MyoD,
myogenin, Id2, and the ribosomal protein S16 that served as an internal control. RNA extracted from mouse SM and brain served as positive
controls. PCR without RT enzyme (R.T) or without cDNA (cDNA) served as negative controls. D, The bands were quantified, and the normalized
values (relative to the control S16 expression) are presented as fold increase. Three independent experiments were conducted and showed similar
kinetic of gene expression.
Mol Endocrinol, January 2011, 25(1):157–169 mend.endojournals.org 159
isolated the 5-flanking region of both genes. Subcloning
mCRFR1 or mCRFR2
5-flanking sequences upstream
to a luciferase gene allowed us to study their activity dur-
ing the myogenic differentiation (Fig. 3). C2C12 cells
were transfected with the reporter plasmid DNA, and the
luciferase activity over3dofmyogenic differentiation
was determined. Interestingly, myoblast differentiation
was accompanied by a significant and rapid inhibition of
the mCRFR1 promoter, and time-dependent and robust
activation of the mCRFR2
promoter (Fig. 3, A and B,
respectively). These sequential changes parallel the de-
crease and the increase in mRNA level of CRFR1 and
CRFR2
, respectively (Fig. 3), and confirm that mCRFR2
expression during differentiation is regulated at the tran-
scriptional level.
To further study the involvement of putative muscle-
specific transcription factors in the activation of the
CRFR2
5-flanking region, the 5-flanking region of
FIG. 2. CRFR1 and CRFR2 signaling pathways are differentially activated during the myogenic differentiation. A, Activation of CRE-luciferase
reporter by CRF and Ucn3 peptides in C2C12 myoblasts or myotubes, respectively. C2C12 myoblasts were transfected with CRE-luciferase, and
luciferase activity was measured after treatment (4 h) with vehicle or 1 pM, 0.1 nM,10nM, and 1
MCRF or Ucn3 in nondifferentiated C2C12 cells
(black bars) or 48 h differentiated C2C12 cells (white bars). Assays were normalized to cotransfected
-gal activity. The mean SEM of three
independent experiments is presented as relative activity. The activation of CRE-luciferase in myoblasts by CRF, or in myotubes by Ucn3, was
blocked by the CRFR1-specific antagonist (Ant) or CRFR2-specific antagonist Ast 2B, respectively [*, P0.05 vs. vehicle treatment; **, P0.05
vs. CRF (10 nM) or Ucn3 (10 nM) treatment]. B. Calcium mobilization in nondifferentiated (myoblasts) or differentiated (myotubes) C2C12 cells by
CRF and Ucn2. Nondifferentiated C2C12 cells (a and c) and differentiated C2C12 (b and d) were treated with CRF (a and b) or Ucn2 (c and d),
respectively, and the calcium mobilization kinetic was measured using FlexStation (Molecular Devices Corp.). Bar graphs represent the maximum
values. Interestingly, CRFR1 but not CRFR2 activation promotes calcium mobilization. Activation of calcium flux was differentially activated in the
myoblasts and myotubes after stimulation with CRF. * P0.05 vs. control buffer. C, CHO cells stably expressing the CRFR1 (a) or CRFR2 (b) were
treated with CRF or Ucn2, respectively, and the calcium mobilization kinetic was measured using FlexStation. Bar graphs represent the maximum
values. *, P0.05 vs. control buffer. ND, Not determined; Con., control.
160 Kuperman et al. Expression and Regulation of Muscle CRFR2
Mol Endocrinol, January 2011, 25(1):157–169
CRFR2
(2495 to 23) was analyzed for SM transcrip-
tion factor consensus sequences using the TESS program
(Transcription Element Search System) (Fig. 4A). Six
fragments of the 5-flanking region, with different
lengths and numbers of putative muscle-specific tran-
scription factors consensus sequences, were subcloned
into a luciferase pGL3 basic vector and used for trans-
fecting C2C12 cells (Fig. 4, B and C). The luciferase
activity of each fragment during myogenic differentia-
tion was studied. No basal differences were detected
between the different fragments. When differentiation
medium (DM) was introduced, the promoter activity
increased in a time-dependant manner. The differences
between the promoter fragments could be detected as
early as 24 h in DM, where the full isolated 5-flanking
region was strongly activated and its activity was sig-
nificantly higher compared with the truncated frag-
ments, regardless of their length (Fig. 4C). After 96 h in
DM, all truncated fragments were robustly activated.
However, their activity was significantly lower com-
pared with the full 5-flanking region (Fig. 4C). Al-
though the activity level of the truncated fragments
varied, there was no significant difference between
them. Interestingly, even the shortest fragment, consist-
ing of 168 bp, was strongly activated, indicating the
importance of the proximal site in mediating CRFR2
transcription (Fig. 4C).
Given the strong potency of the short 5-flanking frag-
ment (146 to 23), we examined the importance of the
putative myocyte enhancer factor (MEF)2 consensus se-
quence (located 91 to 82) for this activation. This MEF2
consensus sequence in the minimal 5-flanking region
was mutated and subcloned into pGL3 basic vector.
The MEF2 consensus sequence is
YTWWAAATAR, where Y stands for
T or C, W stands for A or T, and R
stands for A or G (43). The mutation
included C to A and A to C substitutions
(CtatAaataa to AtatCaataa) (Fig. 5A).
The mutated sequence is not recognized
as MEF2 consensus sequence using the
TESS analysis. The WT or mutated frag-
ment were transfected into C2C12 cells,
and luciferase activity during myogenic
differentiation was measured (Fig. 5B).
The mutated fragment was not able to
induce transcription as demonstrated by
a constant low activation of the lucif-
erase gene throughout the differentia-
tion process, indicating the significance
of this MEF2 site for CRFR2
expres-
sion during myogenic differentiation.
Regulation of SM CRFR2
expression by stress and
its correlation to retinol-binding protein 4 (RBP4)
expression level
Given the high expression level of CRFR2
and its
suggested role in modulating insulin sensitivity and glu-
cose uptake, we further studied its regulation in the ma-
ture mouse after exposure to chronic stressors. High-fat
diet (HFD) and chronic-variable stress (CVS) paradigms
were chosen because they represent prolonged physiolog-
ical and psychological stressors, respectively. SM RNA
obtained from mice maintained for 15 wk on HFD or
from mice subjected to CVS protocol, and the respective
controls, was reverse transcribed, and CRFR2
expres-
sion levels were determined using real-time PCR. Interest-
ingly, both stressors triggered a significant elevation in
CRFR2
expression level. HFD induced a 2.3-fold in-
crease (Fig. 6A) whereas CVS induced a 2.0-fold increase
(Fig. 6B) in CRFR2
expression level. The expression of
Ucn2, the local ligand for SM CRFR2
, did not change
significantly under these conditions (data not shown).
CRFR2 signaling was previously demonstrated to in-
hibit insulin signaling in SM (8). To further understand
the molecular mechanisms mediating the effect of Ucn2/
CRFR2 signaling on insulin sensitivity in SM, we com-
pared the gene expression profile of SM obtained from
both CRFR2 knockout (KO) and WT littermates using
gene expression microarray. The microarray analysis
demonstrated a significant reduction of 43.5% in the ex-
pression level of RBP4. RBP4 is an adipokine whose se-
rum levels are increased in insulin-resistant subjects, and
its administration leads to impaired insulin signaling in
muscle (44). To confirm our microarray data, SM was
FIG. 3. Differential activation of mCRFR1 and mCRFR2
promoters during C2C12 myoblast
differentiation. Schematic demonstration of the mCRFR1 (A) and mCRFR2
(B) 5-flanking
region construct fused to the luciferase gene in PGL3 basic vector. C2C12 cells were
transfected with the reporter plasmid DNA, and luciferase activity during the myogenic
differentiation was determined. The luciferase activity was corrected to
-gal values. (Results
are shown as mean SEM of six independent experiments).
Mol Endocrinol, January 2011, 25(1):157–169 mend.endojournals.org 161
collected from CRFR2-null mice and from their WT litter-
mates, and SM cDNA was used for measuring RBP4 expres-
sion level by real-time PCR. The real-time PCR results were
in agreement with the microarray findings and showed a
significant reduction of 52% in RBP4 expression level (Fig.
7A). Several genes that were found to be up- or down- reg-
ulated in the microarray analysis are listed in Supplemental
Table 1 (published on The Endocrine Society’s Journals
Online web site at http://mend.endojournals.org). These
changes were not further verified using additional quantita-
tive methods. Because CRFR2-KO mice are a developmen-
tal KO model and therefore may represent developmental
compensatory changes, we further examined RBP4 muscle
expression level in conditions that up-regulate CRFR2
ex-
pression, namely HFD and CVS, and found a positive cor-
relation between RBP4 and CRFR2
expression. SM RBP4
FIG. 4. Sequence-, fragmentation-, and differentiation-induced activation of CRFR2
5-flanking region. A, Genomic sequence of mCRFR2
5-
flanking region. 5-Untranslated region is shown in italic letters. The six primers used for promoter fragmentation are underlined, and putative sites
for muscle-specific transcription factors, recognized using TESS (Transcription Element Search System), are indicated. B, Electrophoretic analysis
of pGL3-basic vectors containing truncated mCRFR2
5-flanking region. C, Activity of CRFR2
fragmented promoter during myogenic
differentiation. C2C12 cells were transfected with the reporter plasmid preceded by the different 5-flanking region fragments. Luciferase activity
during the myogenic differentiation in 2% horse serum containing DM was determined. The relative luciferase activity was corrected to
-gal
activity (results are shown as fold increase over the basal activity of each fragment, shown as mean SEM). AS, Antisense.*, P0.05; **, P
0.001 vs. the full fragment.
162 Kuperman et al. Expression and Regulation of Muscle CRFR2
Mol Endocrinol, January 2011, 25(1):157–169
expression level was significantly elevated in mice subjected
to both HFD and CVS manipulations (Fig. 7, B and C).
Discussion
SM tissue has been demonstrated to express high levels of
CRFR2 transcript (20), which was shown to be associated
with controlling glucose transport into the SM. In the
present study we demonstrated, using specific mCRFR2
RNase protection assays, RT-PCR, and DNA sequencing,
that adult SM tissue expresses CRFR2
, but not the
CRFR1, transcripts. Previous reports showed CRFR2
expression in SM to be localized in neural structures,
blood vessels, myotendinous junctions, and endomysial/
perimysial spaces, but not in myocytes (45).
Here, we showed that C2C12 myoblasts exclusively
express CRFR1, whereas the C2C12 myotubes exclu-
sively express CRFR2
. In the myoblast state, serum in-
duces the expression of Id proteins, transcription factors
that sequester E12 and E47 into complexes unable to bind
DNA (39). Upon serum removal, MyoD family proteins,
MyoD, Mrf4, and myogenin, are activated to promote the
expression of muscle-specific genes with Mef2 family of
transcription factors, which play an important role in this
context (46). This sequential expression pattern is also
demonstrated in the CRFR expression kinetics, where
CRFR1 expression mirrors the expression profile of Id2,
and CRFR2
expression kinetics parallels the MyoD and
myogenin expression profile. Additional examples of dif-
ferential expression of CRFRs were reported in other
types of muscle tissues. In human nonpregnant myome-
trium the CRFR1
and CRFR1
-receptor subtypes were
found, whereas at term R2
and C variant CRFR sub-
types were expressed as well (47). Moreover, we recently
reported differential regulation of mCRFR2
by stress in
heart myocardium. The mRNA levels of mCRFR2
were
down-regulated in hearts of mice that underwent CVS
whereas the mRNA levels of a new splice variant of
CRFR2
, iv-mCRFR2
, were up-regulated (22). Lipo-
polysaccharide was also shown to differentially regulate
CRFR2
expression in the heart and SM. Systemic injec-
tion of lipopolysaccharide up-regulated SM CRFR2
mRNA levels and markedly down-regulated its mRNA
heart levels (48).
The pharmacological properties of CRFR1 and
CRFR2 activation by different ligands of the CRF peptide
family are well established. CRF has relatively lower af-
finity for CRFR2 compared with its affinity for CRFR1,
Ucn1 has equal affinities for both receptors, and Ucn2
and Ucn3 appear to be selective for CRFR2 (40, 42, 49).
Both CRFRs belong to the B1 subfamily of seven-trans-
membrane-domain receptors that signal by coupling to G
proteins (50). CRFR1 and CRFR2 signaling primarily
stimulates the adenylyl cyclase/cAMP pathway via cou-
pling and activation of G
s
proteins and protein kinase A
activation (13, 18, 51). In addition, CRFR1 is coupled to
activation of plasma membrane calcium channels and CRF
signaling and was shown to generate changes
in corticotrope cytosolic free calcium concen-
tration (52). The increase in Ca
2
influx in-
volves voltage-gated channels, namely L- and
P-type channels (53). That CRFR1 coupling
to activation of plasma membrane calcium
channels depends on cell type (54) was dem-
onstrated in melanocytes (55). We assessed
the differential expression of CRFR signaling
in assays based on these similarities and dif-
ferences. CRF and Ucn3 activated CRE-
luciferase reporter gene in myoblasts or myo-
tubes, respectively. The use of selective
antagonists and the subsequent activation of
FIG. 6. Up-regulation of SM CRFR2
after exposure to chronic stressors. CRFR2
mRNA level determined by real-time PCR in SM obtained from mice kept on HFD
compared with control low-fat diet (A) or subjected to CVS (B). CRFR2
expression was
corrected by HPRT1 expression level and normalized to control levels (results are shown
as mean SEM). *, P0.05 vs. control.
FIG. 5. Activity of the WT and mutated MEF2 site in the minimal
CRFR2
5-flanking region during the myogenic differentiation. C2C12
cells were transfected with the WT or the mutated minimal (146 to
23) 5-flanking region-luciferase vectors and luciferase activity during
myogenic differentiation in 2% HS containing media (DM) was
determined. The relative luciferase activity was corrected to
-gal
activity (results are shown as mean SEM). Mut, Mutated.
Mol Endocrinol, January 2011, 25(1):157–169 mend.endojournals.org 163
the cAMP pathway further emphasized the ligand specificity
and functionality of the CRFR subtypes. Furthermore, the
ability of CRF to induce calcium mobilization selectively in
myoblasts provides an additional level of support to the
absence of CRFR1 in differentiated myotubes.
Transient transfection of C2C12 myoblasts with con-
structs containing the 5-flanking region of the mCRFR1
or mCRFR2
genes fused to a luciferase reporter showed
differential promoter activity during myogenic differenti-
ation. The CRFR1 promoter activity was negatively reg-
ulated, whereas CRFR2
promoter activity was posi-
tively regulated, during differentiation. This differential
regulation is due to the varying responsiveness of the pro-
moters to myogenic transcription factors. Computer-
aided sequence analysis revealed the presence of putative
muscle-specific transcription factor consensus sequences
in the mCRFR2
5-flanking region. Different fragments
of the 5-flanking region were cloned into a luciferase
vector to identify the crucial area needed for CRFR2
transcription. A robust activation of all the fragmented re-
gions was observed revealing the importance of the 3-prox-
imal region. The importance of this region was verified by
mutating the MEF2 consensus sequence. The mutated frag-
ment was incapable of transcription, as demonstrated by
blunted luciferase activity. This short but powerful minimal
5-flanking region may be further used as a minimal pro-
moter for muscle-specific expression of target genes.
Understanding the regulation of SM CRFR2
may
provide further insight into the physiological functions of
this receptor. Mice lacking either CRFR2 or Ucn2 dem-
onstrate enhanced glucose tolerance, increased insulin
sensitivity, and protection from high fat diet-induced in-
sulin resistance (6, 8). In Ucn2 KO mice, systemic Ucn2
administration before glucose tolerance test or insulin tol-
erance test impaired glucose clearance and reduced insu-
lin sensitivity, respectively (8), showing that this pheno-
type is mediated by peripheral CRFR2. Both obesity and
high stress, hallmarks of a modern lifestyle, are correlated
with insulin resistance (56, 57). We showed that both a
physiological stressor (chronic consumption of a HFD)
and a psychological stressor (CVS)
share the same consequence of ele-
vated SM CRFR2
expression level.
Consequently, the increased SM CRFR2
expression may contribute to the re-
duced insulin sensitivity, which char-
acterizes these conditions. Elucidation
of this phenomenon is essential for bet-
ter management of the metabolic con-
sequences that coincide with both HFD
and chronic psychological stress. Inter-
estingly, a similar CVS protocol medi-
ated a reduction in CRFR2
mRNA lev-
els in the hearts of mice (22); however, these tissue-specific
differences might be attributed to the up-regulation of the
dominant-negative iv-mCRFR2
isoform.
A positive correlation between leptin serum levels and
CRFR2
mRNA levels in the ventromedial hypothala-
mus has been shown (30). Because leptin is produced in
proportion to fat stores (58), and full-length leptin recep-
tor is expressed by SM (59), it is intriguing to hypothesize
that the increased CRFR2
expression under HFD is reg-
ulated by leptin. However, the association with leptin
does not explain the increased CRFR2
expression in
mice subjected to CVS, because CVS was demonstrated to
reduce serum leptin levels (60). Both HFD consumption
and chronic stress lead to elevated glucocorticoids (60
62), which may regulate CRFR2
expression under these
conditions. HFD and CVS may be considered representa-
tives of modern lifestyle characteristics, which include
high stress load and increased intake of high-fat foods.
The indication that mice lacking Ucn2 exhibited in-
creased insulin sensitivity and better glucose tolerance (8)
implies that Ucn2 is endogenously secreted under hypo-
glycemic and hyperglycemic states. Glucose serves as the
primary fuel molecule in the fight or flight response and is
crucial for the organism’s survival (4, 63). Therefore, the
inhibitory effect of SM CRFR2
on insulin signaling may
function to regulate the stress-induced elevation in blood
glucose levels and to allow availability of glucose to other
tissues. Whereas this function is beneficial under normal
conditions, it may be maladaptive under chronic stress
conditions, under which SM CRFR2
expression is ele-
vated and consequently insulin sensitivity is reduced.
Whole-genome microarray expression data comparing
the expression profile of SM obtained from CRFR2 KO
or WT littermates showed a robust reduction in RBP4
expression, which was further confirmed by real-time
PCR. RBP4 is mainly expressed in liver and adipose tissue
(64). RBP4 serum levels are increased in insulin-resistant
mice and in humans with type 2 diabetes (65), and weight
loss in morbidly obese patients reduces RBP4 serum level
FIG. 7. SM RBP4 and CRFR2
expression levels are positively correlated. RBP4 mRNA level
determined by real-time PCR in SM obtained from CRFR2 KO mice and their WT littermates
(A), mice kept on HFD (B) or mice subjected to CVS (C). RBP4 expression was corrected by
HPRT1 expression level and normalized to WT/control (results are shown as mean SEM).
*, P0.05 vs. WT/control.
164 Kuperman et al. Expression and Regulation of Muscle CRFR2
Mol Endocrinol, January 2011, 25(1):157–169
(66). Adipose-specific glucose transporter 4 KO mice
demonstrated elevated serum RBP4 levels and secondary
insulin resistance in the muscle and the liver (65), a met-
abolic phenotype that mirrors the observed phenotype of
the CRFR2 and Ucn2 KO mice.
RBP4 and CRFR2 signaling disrupt components of SM
insulin signaling that play a role in the control of glucose
transporter 4 translocation. RBP4 reduces both phospho-
inositide3-kinase activity and insulin-stimulated tyrosine
phosphorylation of insulin receptor substrate-1 at ty-
rosine residue 612, a docking site for the p85 subunit of
phosphoinositide3-kinase (65), whereas Ucn2 signaling
inhibits insulin-induced Akt phosphorylation and reduces
ERK1/2 phosphorylation (8). It was demonstrated that
RBP expressed ectopically in mice muscle can elevate se-
rum RBP levels (65). Here, we demonstrate that HFD and
CVS conditions mediate an increase both in SM CRFR2
and RBP4 expression levels. This dual increased expression
may synergistically act in an autocrine fashion to inhibit
insulin signaling and magnify metabolic complications.
The current findings further position the SM-CRFR2
pathways as a relevant physiological system that may af-
fect the known reciprocal relationship between psycho-
logical and physiological challenges and the metabolic
syndrome. A better understanding of SM CRFR2
path-
way, its physiological roles, and its regulation may pro-
vide benefits in related pathological conditions, such as
obesity and type 2 diabetes.
Materials and Methods
Animals
Mice were housed and handled in a pathogen-free temperature-
controlled (22 C 1) mouse facility on a 12-h light, 12-h dark cycle
(lights on from 1900 h–0700 h), with food and water given ad
libitum, according to institutional guidelines. Adult C57BL/6 male
mice were used in all experiments. CRFR2-null (129C57Bl/6
mixed background) mice were used for microarray and real-time
PCR studies. All experimental protocols were approved by the
Institutional Animal Care and Use Committee of The Weizmann
Institute of Science.
Cell lines
C2C12 myoblasts were grown to 50% confluence in DMEM
(Invitrogen Life Technologies, Carlsbad, CA), containing 10%
FBS supplemented with 100
g/ml of penicillin/streptomycin
(Invitrogen Life Technologies) (normal growth medium). For
differentiation, C2C12 were grown to 90% confluency and
washed with serum-free medium, and their medium was replaced
with DMEM containing 2% horse serum. CHO cells were grown
in normal growth medium as previously described (22).
RNA and cDNA preparation
RNA was extracted from brain, pituitary, gastrocnemius
muscle, or C2C12 cells using Tri-Reagent RNA isolation re-
agent (Molecular Research Center, Cincinnati, OH) according
to the manufacturer’s recommendations. To avoid false-positive
results caused by DNA contamination, a deoxyribonuclease
treatment was performed for 30 min at 37 C using the RQ1 RNase-
free deoxyribonuclease (Promega Corp., Madison, WI). RNA
preparations were reverse transcribed to generate cDNA using
High Capacity cDNA Reverse Transcription Kit (Applied Biosys-
tems, Inc., Foster city, CA). The cDNA products were used as
templates for semiquantitative and quantitative PCR analysis.
Semiquantitative RT-PCR
Semiquantitative RT-PCR was used to amplify the levels of
endogenous mCRFR2 and mCRFR1 present in the mouse SM and
brain. The expression of mCRFR1, mCRFR2 as well as muscle
differentiation markers MyoD, myogenin and Id2 levels were stud-
ied during C2C12 differentiation. The cDNA products were used
as templates for semiquantitative RT-PCR analysis using specific
primers for mCRFR2, mCRFR1, MyoD, myogenin, and Id2 and
the ribosomal protein S16 (for sequences see Table 1).
TABLE 1. Sequence of PCR primers
Gene Primer sequence (5to 3) GenBank accession no.
CRFR1 NM_007762
Sense GGT GTG CCT TTC CCC ATC ATT
Antisense CAA CAT GTA GGT GAT GCC CAG
CRFR2 NM_009953
Sense GGC AAG GAA GT GGT GAT TTG
Antisense GGC GTG GTG GTC CTG CCA GCG
MyoD1 NM_010866
Sense GAG CAA AGT GAA TGA GGC CTT
Antisense CAC TGT AGT AGG CGG TGT CGT
Myogenin NM_031189
Sense TCA GAA GAG GAT GCT CTC TGC
Antisense TCA GAA GAG GAT GCT CTC TGC
Id2 NM_010496
Sense ATG AAA GCC TTC AGT CCG GTG
Antisense TTA GCC ACA GAG TAC TTT GCT
S16 M11408
Sense TGC GGT GTG GAG CTC GTG CTT GT
Antisense GCT ACC AGG CCT TTG AGA TGG A
Mol Endocrinol, January 2011, 25(1):157–169 mend.endojournals.org 165
PCR without reverse transcriptase enzyme (R.T) or with-
out cDNA (cDNA) served as negative control. The expression
of ribosomal protein S16 served as internal control. The PCR
conditions were as follows: cDNA equivalent to 200 ng of total
RNA was amplified by PCR for 35 cycles at an annealing tem-
perature of 62 C. The final MgCl
2
concentration was 3 mM, and
each reaction contained 2.5 U of Taq DNA polymerase (BIO-
X-ACT DNA polymerase; Bioline UK Ltd., London, UK).
RNase protection assay
SM total RNA was hybridized with the mCRFR1, mCRFR2,
and glyceraldehyde-3-phosphate dehydrogenase antisense probes.
Brain and pituitary gland served as positive control for CRFR2 and
CRFR1 gene expression, respectively. RNase protection assay was
performed as previously described (23).
Transient transfections and luciferase assay
C2C12 were used for the CRE activation and for the pro-
moter studies. All transfections were carried out in 12-well
plates using Lipofectamine 2000 Transfection Reagent (Invitro-
gen Life Technologies) according to manufacturer’s instruc-
tions. For CRE-luciferase activation, C2C12 myoblasts were
plated to 90% confluency and transfected with 1.5
gofthe
luciferase reporter containing a fragment of the EVX1 gene,
which contains a potent CRE site (kindly provided by Marc
Montminy, The Salk Institute) and 50 ng
-gal expression plas-
mid. Cells were treated for 4 h with vehicle or 1 pM, 0.1 nM,10
nM, and 1
MCRF or Ucn3 in nondifferentiated C2C12 cells, or
48 h differentiated C2C12 cells with or without the presence of
CRFR1- and CRFR2-selective antagonists (Ant and Ast 2B, re-
spectively). For promoter studies, C2C12 myoblasts were plated
to 90% confluency and transfected with 1.5
g of the luciferase
reporter plasmid or empty pGL3 vector and 50 ng
-gal expres-
sion plasmid. After 24 h the medium was replaced with DM.
The promoter activity was monitored at the basal state and after
the indicated times (24–96 h) in DM. The cells were harvested,
and the luciferase reporter activity was assayed as previously
described (20). Transfections were performed at least three
times (in triplicate) for each construct or treatment tested. To
correct for variations in transfection efficiencies, luciferase ac-
tivities were normalized to
-gal activity. Results were corrected
by the activity of the promoterless pGL3 vector.
Calcium-mobilization assay
Calcium-mobilization kinetics in nondifferentiated or differ-
entiated C2C12 cells or in CHO cells stably transfected with
either mCRFR1 or mCRFR2 after treatment of CRF or Ucn2
(50 nM) were measured using FlexStation (Molecular Devices,
Sunnyvale, CA) as previously described (67).
Construction of luciferase reporter plasmids
The mCRFR1 and mCRFR2
5-flanking region constructs
were cloned by PCR using mouse genomic DNA. The primers
used for the construct were designed to include artificial restric-
tion sites (KpnI and XhoI for mCRFR1; KpnI and NheI for
mCRFR2
). The primer sequences were as follows: for
mCRFR1 sense primer (2685 to 2663): 5-TTG GGT TAC
GTA TGC TGC TCC TT-3and antisense primer (196 to
217): 5-CCT CGG GCT CGC TCT GTC AGC-3. For
mCRFR2
sense primer (2495 to 2473): 5-GGA AAT GCA
GGA AAG CCA AGA CA-3and antisense primer (4to23):
5-CTG CCC GAC CTA CCC ACC AA-3. Fragmentation of
the mCRFR2
5-flanking region was done using the above
mentioned antisense primer along with six sense primers located
at: (1649 to 1630), (1028 to 1009), (955 to 932),
(747 to 726), (512 to 492), (146 to 127). The prim-
ers sequences are indicated in Fig. 4; all primers contained an
artificial KpnI restriction site. For mutating the MEF2 recogni-
tion site located at 91 to 82, the proximal 5-flanking area
was amplified using the above-mentioned antisense primer with
the following sense primer, which contains an endogenous PstI
restriction site: 5-CTGCAGAAGTTGCTGCCCAGAGCCA-
GATATCAATAACCTGG-3. The mutation introduces a
unique EcoRV restriction site (in italics), which was later used
for identifying mutated clones. The PCR products were ana-
lyzed by agarose gel electrophoresis and eluted from the gel.
After digestion by the appropriate restriction enzymes, the DNA
fragments were cloned into the luciferase reporter plasmid
pGL3 (Promega Corp.), and the sequences were verified using
automated direct DNA sequencing.
HFD
Mice were fed ad libitum a high-fat (60% of calories) (n
13) or low-fat (10% of calories) (n 5) diet (D12492 and
D12450B, respectively; Research Diets, Inc., New Brunswick,
NJ) for 15 wk.
CVS
CVS mice (n 5) were housed in a temperature-controlled
room (22 C 1) and were subjected to the CVS protocol for a
period of 4 wk as previously described (22).
Real-Time PCR
SM cDNA products were used as templates for real-time PCR
analysis. Sense and antisense primers were selected to be located on
different exons to avoid false-positive results caused by DNA con-
tamination. The following specific primers were designed using
Primer Express software (Applied Biosystems, PerkinElmer, Foster
City, CA). For mCRFR2: 5-TACCGAATCGCCCTCATTGT-3
and 5-CCACGCGATGTTTCTCAGAAT-3corresponding to
nucleotides 479-498 and 640-620, respectively (GenBank acces-
sion no. AY445512); for mRBP4: 5-GCTTCCGAGTCAAG-
GAGAACTTC-3and 5-TCCACAGAAAACTCAGC-
GATGA-3corresponding to nucleotides 479-498 and 640-620,
respectively (GenBank accession no. NM_011255). For mouse
hypoxanthine guanine phosphoribosyl transferase 1 (HPRT1),
which served as an internal control: 5-GCAGTACAGC-
CCCAAAATGG-3and 5-GGTCCTTTTCACCAG-
CAAGCT-3corresponding to nucleotides 389-411 and 509-488,
respectively (GenBank accession no. NM_013556). Real-time PCRs
were carried out on a 7500 Real-Time PCR system (Applied Bio-
systems, Inc.), using fluorescent SYBR Green technology (Abgene;
Epsom, Surrey, UK). Reaction protocols had the following format:
15 min at 95 C for enzyme activation, followed by 45 cycles of 15
sec at 94 C and 60 sec at 60 C. The specificity of the amplification
products was checked by melting curve analysis. All reactions con-
tained the same amount of cDNA, 10
l Master Mix, and 250 nM
primers to a final volume of 20
l.
Microarray preparation and data are described under Gene
Expression Omnibus accession number GSE25045. Briefly, to-
tal RNA was extracted from adult skeletal muscle obtained
from four CRFR2 KO mice and four WT littermates, using
Tri-Reagent RNA isolation reagent (Molecular Research Cen-
166 Kuperman et al. Expression and Regulation of Muscle CRFR2
Mol Endocrinol, January 2011, 25(1):157–169
ter, Cincinnati, OH) according to the manufacturer’s protocol.
The RNA was pooled such that each sample consisted of two
muscles from each genotype (a total of four samples). Total
RNA (100
g) was further cleaned using Qiagen RNA purifi-
cation kit (QIAGEN Inc., Valencia, CA), and RNA integrity was
verified using gel electrophoresis and 260/280 ratios. cRNA
synthesis and hybridization to Affymetrix Murine Genome-
U74Av2 array (Affymetrix, Santa Clara, CA) was performed by
the UCSD Biological Services Unit. Data was analyzed using
Affymetrix Microarray Analysis Suite 5.1.
Acknowledgments
We thank Mr. S. Ovadia for his devoted assistance with animal
care.
This research was supported in part by the Clayton Medical
Research Foundation, Inc. A.C. was supported by the following:
Roberto and Renata Ruhman, Brazil; Mark Besen and the Pratt
Foundation, Australia; the Israel Science Foundation; the Leg-
acy Heritage Biomedical Science Partnership D-Cure Fellow-
ship; Nella and Leon Benoziyo Center for Neurosciences; Nella
and Leon Benoziyo Center for Neurological Diseases; Carl and
Micaela Einhorn-Dominic Brain Research Institute; Irwin
Green Alzheimer’s Research Fund; Gerhard and Hannah Ba-
charach (Fort Lee, NJ) and is incumbent of the Philip Harris and
Gerald Ronson Career Development Chair. W.V. was sup-
ported by Award No. 5P01DK026741-30 from the National
Institute of Diabetes and Digestive and Kidney Diseases and is a
Clayton Medical Research Foundation, Inc. Senior Investigator
and the Helen McLoraine Professor of Molecular Neurobiology.
Address all correspondence and requests for reprints to:
Alon Chen, Ph.D., Department of Neurobiology, Weizmann
Institute of Science, Rehovot, Israel 76100. E-mail: alon.
chen@weizmann.ac.il.
Disclosure Summary: W.V. is a cofounder, consultant, equity
holder, and member of the Board of Directors of Neurocrine
Biosciences, Inc., and Acceleron Pharma, Inc. These companies
are developing products that are related to some of the topics
discussed. However, none if these products are as yet on the
market. The content is solely the responsibility of the authors
and does not necessarily represent the official views of the Na-
tional Institute of Diabetes and Digestive and Kidney Diseases
or the National Institutes of Health. Y.K., O.I., J.V., L.B., and
A.C. have nothing to disclose.
References
1. Petersen KF, Shulman GI 2002 Pathogenesis of skeletal muscle insulin
resistance in type 2 diabetes mellitus. Am J Cardiol 90:11G–18G
2. Petersen KF, Dufour S, Savage DB, Bilz S, Solomon G, Yonemitsu S,
Cline GW, Befroy D, Zemany L, Kahn BB, Papademetris X, Rothman
DL, Shulman GI 2007 The role of skeletal muscle insulin resistance
in the pathogenesis of the metabolic syndrome. Proc Natl Acad Sci
USA 104:12587–12594
3. Shulman GI 2004 Unraveling the cellular mechanism of insulin
resistance in humans: new insights from magnetic resonance spec-
troscopy. Physiology (Bethesda) 19:183–190
4. Brown MR, Fisher LA, Spiess J, Rivier C, Rivier J, Vale W 1982
Corticotropin-releasing factor: actions on the sympathetic nervous
system and metabolism. Endocrinology 111:928–931
5. Dallman MF, Akana SF, Strack AM, Hanson ES, Sebastian RJ 1995
The neural network that regulates energy balance is responsive to
glucocorticoids and insulin and also regulates HPA axis responsivity at
a site proximal to CRF neurons. Ann NY Acad Sci 771:730–742
6. Bale TL, Anderson KR, Roberts AJ, Lee KF, Nagy TR, Vale WW
2003 Corticotropin-releasing factor receptor-2-deficient mice dis-
play abnormal homeostatic responses to challenges of increased
dietary fat and cold. Endocrinology 144:2580–2587
7. Carlin KM, Vale WW, Bale TL 2006 Vital functions of corticotrop-
in-releasing factor (CRF) pathways in maintenance and regulation
of energy homeostasis. Proc Natl Acad Sci USA 103:3462–3467
8. Chen A, Brar B, Choi CS, Rousso D, Vaughan J, Kuperman Y, Kim
SN, Donaldson C, Smith SM, Jamieson P, Li C, Nagy TR, Shulman
GI, Lee KF, Vale W 2006 Urocortin 2 modulates glucose utilization
and insulin sensitivity in skeletal muscle. Proc Natl Acad Sci USA
103:16580–16585
9. Kuperman Y, Chen A 2008 Urocortins: emerging metabolic and en-
ergy homeostasis perspectives. Trends Endocrinol Metab 19:122–129
10. Huising MO, van der Meulen T, Vaughan JM, Matsumoto M,
Donaldson CJ, Park H, Billestrup N, Vale WW 2010 CRFR1 is
expressed on pancreatic
cells, promotes
cell proliferation, and
potentiates insulin secretion in a glucose-dependent manner. Proc
Natl Acad Sci USA 107:912–917
11. Kuperman Y, Issler O, Regev L, Musseri I, Navon I, Neufeld-Cohen
A, Gil S, Chen A 2010 Perifornical urocortin-3 mediates the link
between stress-induced anxiety and energy homeostasis. Proc Natl
Acad Sci USA 107:8393–8398
12. Vita N, Laurent P, Lefort S, Chalon P, Lelias JM, Kaghad M, Le Fur
G, Caput D, Ferrara P 1993 Primary structure and functional ex-
pression of mouse pituitary and human brain corticotrophin releas-
ing factor receptors. FEBS Lett 335:1–5
13. Chen R, Lewis KA, Perrin MH, Vale WW 1993 Expression cloning
of a human corticotropin-releasing-factor receptor. Proc Natl Acad
Sci USA 90:8967–8971
14. Chang CP, Pearse II RV, O’Connell S, Rosenfeld MG 1993 Identi-
fication of a seven transmembrane helix receptor for corticotropin-
releasing factor and sauvagine in mammalian brain. Neuron 11:
1187–1195
15. Stenzel P, Kesterson R, Yeung W, Cone RD, Rittenberg MB,
Stenzel-Poore MP 1995 Identification of a novel murine receptor
for corticotropin-releasing hormone expressed in the heart. Mol
Endocrinol 9:637–645
16. Kishimoto T, Pearse II RV, Lin CR, Rosenfeld MG 1995 A sauvag-
ine/corticotropin-releasing factor receptor expressed in heart and
skeletal muscle. Proc Natl Acad Sci USA 92:1108–1112
17. Perrin M, Donaldson C, Chen R, Blount A, Berggren T, Bilezikjian
L, Sawchenko P, Vale W 1995 Identification of a second cortico-
tropin-releasing factor receptor gene and characterization of a
cDNA expressed in heart. Proc Natl Acad Sci USA 92:2969–2973
18. Lovenberg TW, Liaw CW, Grigoriadis DE, Clevenger W, Chalmers
DT, De Souza EB, Oltersdorf T 1995 Cloning and characterization
of a functionally distinct corticotropin-releasing factor receptor
subtype from rat brain. Proc Natl Acad Sci USA 92:836–840
19. Van Pett K, Viau V, Bittencourt JC, Chan RK, Li HY, Arias C, Prins
GS, Perrin M, Vale W, Sawchenko PE 2000 Distribution of mRNAs
encoding CRF receptors in brain and pituitary of rat and mouse.
J Comp Neurol 428:191–212
20. Chen A, Perrin M, Brar B, Li C, Jamieson P, Digruccio M, Lewis K,
Vale W 2005 Mouse corticotropin-releasing factor receptor type
2alpha gene: isolation, distribution, pharmacological characteriza-
tion and regulation by stress and glucocorticoids. Mol Endocrinol
19:441–458
21. Lovenberg TW, Chalmers DT, Liu C, De Souza EB 1995 CRF2
and CRF2
receptor mRNAs are differentially distributed between
the rat central nervous system and peripheral tissues. Endocrinol-
ogy 136:4139–4142
Mol Endocrinol, January 2011, 25(1):157–169 mend.endojournals.org 167
22. Sztainberg Y, Kuperman Y, Issler O, Gil S, Vaughan J, Rivier J, Vale
W, Chen A 2009 A novel corticotropin-releasing factor receptor
splice variant exhibits dominant negative activity: a putative link to
stress-induced heart disease. FASEB J 23:2186–2196
23. Chen A, Blount A, Vaughan J, Brar B, Vale W 2004 Urocortin II
gene is highly expressed in mouse skin and skeletal muscle tissues:
localization, basal expression in corticotropin-releasing factor re-
ceptor (CRFR) 1- and CRFR2-null mice, and regulation by glu-
cocorticoids. Endocrinology 145:2445–2457
24. Hall JE, Kaczor JJ, Hettinga BP, Isfort RJ, Tarnopolsky MA 2007
Effects of a CRF2R agonist and exercise on mdx and wildtype
skeletal muscle. Muscle Nerve 36:336–341
25. Hinkle RT, Donnelly E, Cody DB, Bauer MB, Isfort RJ 2003 Uro-
cortin II treatment reduces skeletal muscle mass and function loss
during atrophy and increases nonatrophying skeletal muscle mass
and function. Endocrinology 144:4939–4946
26. Brar BK, Jonassen AK, Egorina EM, Chen A, Negro A, Perrin MH,
Mjos OD, Latchman DS, Lee KF, Vale W 2004 Urocortin-II and
urocortin-III are cardioprotective against ischemia reperfusion in-
jury: an essential endogenous cardioprotective role for cortico-
tropin releasing factor receptor type 2 in the murine heart. Endo-
crinology 145:24–35; discussion 21–23
27. Makino S, Asaba K, Takao T, Hashimoto K 1998 Type 2 cortico-
tropin-releasing hormone receptor mRNA expression in the heart
in hypertensive rats. Life Sci 62:515–523
28. Makino S, Nishiyama M, Asaba K, Gold PW, Hashimoto K 1998
Altered expression of type 2 CRH receptor mRNA in the VMH by
glucocorticoids and starvation. Am J Physiol 275:R1138–R1145
29. Makino S, Asaba K, Nishiyama M, Hashimoto K 1999 Decreased
type 2 corticotropin-releasing hormone receptor mRNA expression
in the ventromedial hypothalamus during repeated immobilization
stress. Neuroendocrinology 70:160–167
30. Nishiyama M, Makino S, Asaba K, Hashimoto K 1999 Leptin
effects on the expression of type-2 CRH receptor mRNA in the
ventromedial hypothalamus in the rat. J Neuroendocrinol 11:307–
314
31. Eghbal-Ahmadi M, Avishai-Eliner S, Hatalski CG, Baram TZ 1999
Differential regulation of the expression of corticotropin-releasing
factor receptor type 2 (CRF2) in hypothalamus and amygdala of the
immature rat by sensory input and food intake. J Neurosci 19:
3982–3991
32. Kageyama K, Gaudriault GE, Bradbury MJ, Vale WW 2000 Reg-
ulation of corticotropin-releasing factor receptor type 2
messen-
ger ribonucleic acid in the rat cardiovascular system by urocortin,
glucocorticoids, and cytokines. Endocrinology 141:2285–2293
33. Kageyama K, Gaudriault GE, Suda T, Vale WW 2003 Regulation
of corticotropin-releasing factor receptor type 2
mRNA via cyclic
AMP pathway in A7r5 aortic smooth muscle cells. Cell Signal 15:
17–25
34. Coste SC, Heldwein KA, Stevens SL, Tobar-Dupres E, Stenzel-
Poore MP 2001 IL-1
and TNF
down-regulate CRH receptor-2
mRNA expression in the mouse heart. Endocrinology 142:3537–
3545
35. Pournajafi-Nazarloo H, Partoo L, Sanzenbacher L, Azizi F, Carter
CS 2007 Modulation of corticotropin-releasing hormone type 2
receptor and urocortin 1 and urocortin 2 mRNA expression in the
cardiovascular system of prairie voles following acute or chronic
stress. Neuroendocrinology 86:17–25
36. Biressi S, Molinaro M, Cossu G 2007 Cellular heterogeneity during
vertebrate skeletal muscle development. Dev Biol 308:281–293
37. Puri PL, Bhakta K, Wood LD, Costanzo A, Zhu J, Wang JY 2002 A
myogenic differentiation checkpoint activated by genotoxic stress.
Nat Genet 32:585–593
38. Kislinger T, Gramolini AO, Pan Y, Rahman K, MacLennan DH,
Emili A 2005 Proteome dynamics during C2C12 myoblast differ-
entiation. Mol Cell Proteomics 4:887–901
39. Puri PL, Sartorelli V 2000 Regulation of muscle regulatory factors
by DNA-binding, interacting proteins, and post-transcriptional
modifications. J Cell Physiol 185:155–173
40. Reyes TM, Lewis K, Perrin MH, Kunitake KS, Vaughan J, Arias
CA, Hogenesch JB, Gulyas J, Rivier J, Vale WW, Sawchenko PE
2001 Urocortin II: a member of the corticotropin-releasing factor
(CRF) neuropeptide family that is selectively bound by type 2 CRF
receptors. Proc Natl Acad Sci USA 98:2843–2848
41. Hsu SY, Hsueh AJ 2001 Human stresscopin and stresscopin-related
peptide are selective ligands for the type 2 corticotropin-releasing
hormone receptor. Nat Med 7:605–611
42. Lewis K, Li C, Perrin MH, Blount A, Kunitake K, Donaldson C,
Vaughan J, Reyes TM, Gulyas J, Fischer W, Bilezikjian L, Rivier J,
Sawchenko PE, Vale WW 2001 Identification of urocortin III, an
additional member of the corticotropin-releasing factor (CRF) fam-
ily with high affinity for the CRF2 receptor. Proc Natl Acad Sci USA
98:7570–7575
43. Yu YT, Breitbart RE, Smoot LB, Lee Y, Mahdavi V, Nadal-Ginard
B1992 Human myocyte-specific enhancer factor 2 comprises a
group of tissue-restricted MADS box transcription factors. Genes
Dev 6:1783–1798
44. Graham TE, Yang Q, Blu¨ her M, Hammarstedt A, Ciaraldi TP,
Henry RR, Wason CJ, Oberbach A, Jansson PA, Smith U, Kahn BB
2006 Retinol-binding protein 4 and insulin resistance in lean, obese,
and diabetic subjects. N Engl J Med 354:2552–2563
45. Samuelsson S, Lange JS, Hinkle RT, Tarnopolsky M, Isfort RJ 2004
Corticotropin-releasing factor 2 receptor localization in skeletal
muscle. J Histochem Cytochem 52:967–977
46. Buckingham M, Bajard L, Chang T, Daubas P, Hadchouel J, Mei-
lhac S, Montarras D, Rocancourt D, Relaix F 2003 The formation
of skeletal muscle: from somite to limb. J Anat 202:59–68
47. Grammatopoulos D, Dai Y, Chen J, Karteris E, Papadopoulou N,
Easton AJ, Hillhouse EW 1998 Human corticotropin-releasing hor-
mone receptor: differences in subtype expression between pregnant
and nonpregnant myometria. J Clin Endocrinol Metab 83:2539
2544
48. Heldwein KA, Duncan JE, Stenzel P, Rittenberg MB, Stenzel-Poore
MP 1997 Endotoxin regulates corticotropin-releasing hormone re-
ceptor 2 in heart and skeletal muscle. Mol Cell Endocrinol 131:
167–172
49. Vaughan J, Donaldson C, Bittencourt J, Perrin MH, Lewis K,
Sutton S, Chan R, Turnbull AV, Lovejoy D, Rivier C, et al. 1995
Urocortin, a mammalian neuropeptide related to fish urotensin I
and to corticotropin-releasing factor. Nature 378:287–292
50. Bale TL, Vale WW 2004 CRF and CRF receptors: role in stress
responsivity and other behaviors. Annu Rev Pharmacol Toxicol
44:525–557
51. Perrin MH, Vale WW 1999 Corticotropin releasing factor recep-
tors and their ligand family. Ann NY Acad Sci 885:312–328
52. Gue´ rineau N, Corcuff JB, Tabarin A, Mollard P 1991 Spontaneous
and corticotropin-releasing factor-induced cytosolic calcium tran-
sients in corticotrophs. Endocrinology 129:409–420
53. Kuryshev YA, Childs GV, Ritchie AK 1996 Corticotropin-releasing
hormone stimulates Ca2entry through L- and P-type Ca2chan-
nels in rat corticotropes. Endocrinology 137:2269–2277
54. Hauger RL, Olivares-Reyes JA, Braun S, Catt KJ, Dautzenberg FM
2003 Mediation of corticotropin releasing factor type 1 receptor
phosphorylation and desensitization by protein kinase C: a possible
role in stress adaptation. J Pharmacol Exp Ther 306:794–803
55. Fazal N, Slominski A, Choudhry MA, Wei ET, Sayeed MM 1998
Effect of CRF and related peptides on calcium signaling in human
and rodent melanoma cells. FEBS Lett 435:187–190
56. Kyrou I, Tsigos C 2007 Stress mechanisms and metabolic compli-
cations. Horm Metab Res 39:430–438
57. James WP, Rigby N, Leach R 2006 Obesity and the metabolic
syndrome: the stress on society. Ann NY Acad Sci 1083:1–10
58. Mu¨ nzberg H, Myers Jr MG 2005 Molecular and anatomical deter-
minants of central leptin resistance. Nat Neurosci 8:566–570
168 Kuperman et al. Expression and Regulation of Muscle CRFR2
Mol Endocrinol, January 2011, 25(1):157–169
59. Liu YL, Emilsson V, Cawthorne MA 1997 Leptin inhibits glycogen
synthesis in the isolated soleus muscle of obese (ob/ob) mice. FEBS
Lett 411:351–355
60. Lu XY, Kim CS, Frazer A, Zhang W 2006 Leptin: a potential novel
antidepressant. Proc Natl Acad Sci USA 103:1593–1598
61. Dallman MF, Pecoraro N, Akana SF, La Fleur SE, Gomez F,
Houshyar H, Bell ME, Bhatnagar S, Laugero KD, Manalo S 2003
Chronic stress and obesity: a new view of “comfort food.” Proc
Natl Acad Sci USA 100:11696–11701
62. Dallman MF, Akana SF, Strack AM, Scribner KS, Pecoraro N, La
Fleur SE, Houshyar H, Gomez F 2004 Chronic stress-induced ef-
fects of corticosterone on brain: direct and indirect. Ann NY Acad
Sci 1018:141–150
63. Ulrich-Lai YM, Herman JP 2009 Neural regulation of endocrine
and autonomic stress responses. Nat Rev Neurosci 10:397–409
64. Blaner WS 1989 Retinol-binding protein: the serum transport pro-
tein for vitamin A. Endocr Rev 10:308–316
65. Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, Zabolotny
JM, Kotani K, Quadro L, Kahn BB 2005 Serum retinol binding
protein 4 contributes to insulin resistance in obesity and type 2
diabetes. Nature 436:356–362
66. Haider DG, Schindler K, Prager G, Bohdjalian A, Luger A, Wolzt
M, Ludvik B 2007 Serum retinol-binding protein 4 is reduced after
weight loss in morbidly obese subjects. J Clin Endocrinol Metab
92:1168–1171
67. McDermott DH, Fong AM, Yang Q, Sechler JM, Cupples LA,
Merrell MN, Wilson PW, D’Agostino RB, O’Donnell CJ, Patel DD,
Murphy PM 2003 Chemokine receptor mutant CX3CR1-M280
has impaired adhesive function and correlates with protection from
cardiovascular disease in humans. J Clin Invest 111:1241–1250
JCEM includes valuable patient information
from The Hormone Foundation!
www.endo-society.org
Mol Endocrinol, January 2011, 25(1):157–169 mend.endojournals.org 169
... In addition to ghrelin isoforms, the corticotropin-releasing factor (CRF) family of peptides and receptors (Chen et al., 1993;Lovenberg et al., 1995;Perrin et al., 1995;Vale et al., 1981) has also been shown to control appetite and skeletal muscle energy balance. Levels of CRF receptor type 2 (CRF-R2) are elevated by high-fat feeding and chronic variable stress conditions in skeletal muscle (Kuperman et al., 2011). These conditions are also related to muscle insulin resistance (Corcoran et al., 2007;Hung & Ikizler, 2011;Kewalramani et al., 2010;Li et al., 2013;Martins et al., 2012;Mei et al., 2011). ...
... This result contradicts our data, a discrepancy that may be attributed to the use of myotubes, which are well-differentiated muscle cells, in Gortan Cappellari et al.'s (2016) study, whereas we used C2C12 myoblasts. The fact that C2C12 myoblasts exclusively express CRF-R1, whereas differentiated C2C12 myotubes primarily express CRF-R2 (Kuperman et al., 2011), combined with our finding that des-acyl ghrelin significantly upregulates CRF-R1 levels (Gershon & Vale, 2014), support the suggested explanation and also raise the possibility that des-acyl ghrelin upregulates the expression of CRF-R1 in C2C12 cells. ...
... Our published study (Gershon & Vale, 2014) and the data presented here support the notion that CRF-R2 is a downstream effector of ghrelin and des-acyl ghrelin which might be involved in mediating both ghrelin isoforms' effects on muscle metabolism. The ability of CRF-R antagonists to block the effects of des-acyl ghrelin on RBP4 expression in C2C12 cells provides additional support for02 the relationship between CRF-Rs and ghrelin, as does a study showing that skeletal muscles isolated from CRF-R2-null mice express significantly less RBP4 compared to the wild type (Kuperman et al., 2011). ...
Article
Full-text available
Ghrelin and the corticotropin-releasing factor (CRF) family are known regulators of cellular metabolism and energy balance. We previously demonstrated that myoblast glucose metabolism is regulated by ghrelin and that this effect is mediated by CRF receptor type 2 (CRF-R2). Here we explored the effect of des-acyl ghrelin, the major circulating isoform of ghrelin, on cellular metabolism in mouse myoblast C2C12 cells, and examined whether CRF family receptors mediate its metabolic effects in muscle cells. C2C12 cells were exposed to des-acyl ghrelin with or without the CRF-R1- and CRF-R2-specific antagonists antalarmin or antisauvagine-30, respectively. Des-acyl ghrelin reduced glucose uptake and expression of the glucose transporter GLUT4, but induced retinol-binding protein 4 (RBP4) expression. Antalarmin and antisauvagine-30 inhibited the induction of glucose uptake by des-acyl ghrelin and its effect on GLUT4 and RBP4 expression. Moreover, treating C2C12 cells with des-acyl ghrelin resulted in cAMP activation in response to the CRF-R1-specific ligand stressin, and the CRF-R2-specific ligand Ucn3. Furthermore, des-acyl ghrelin reduced the expression of uncoupling proteins UCP2 and UCP3. Adding antalarmin or antisauvagine-30 to the medium reversed this effect. Finally, des-acyl ghrelin elevated lipid content and acetyl-CoA carboxylase expression in C2C12 cells. Our results suggest that during food deprivation, des-acyl ghrelin signals the muscle cells that glucose levels are low and that they should switch to fatty acids for their metabolic fuel.
... CRF2R is a G as protein-coupled receptor (GPCR) that can activate adenylyl cyclase (AC), which enhances intracellular cAMP levels, and consequently activates cAMP-dependent protein kinase (PKA) [5,13]. Indeed, Ucn2 has been shown to stimulate the in vitro production of cAMP in C2C12 myotubes [17] and in isolated mouse muscles [14,15]. Cyclic AMP enhancers such as the nonspecific phosphodiesterase (PDE) inhibitor isobutylmethylxanthine and the selective PDE4 inhibitor rolipram have shown to suppress muscle protein degradation [18]. ...
... The Ucn2 mRNA increase was accompanied by a higher content of Ucn2 protein ( Figure 1B), which mainly remained around myofibers as compared with the intracellular medium, suggesting that this peptide may be synthesized and secreted by myocytes and exerts its autocrine/paracrine effects on muscle cells. Since it is well known that Ucn2 increases cAMP levels in vitro [14,15,17] we confirmed that in vivo in Ucn2-transfected TA muscles for 14 days (1508 AE 109* vs 1082 AE 100 cAMP fmol/mg of muscle, control group; n ¼ 3). Then, the involvement of PKA, the canonical downstream effector of cAMP, was investigated. ...
Article
Full-text available
Objective Although it is well established that urocortin 2 (Ucn2), a peptide member of the corticotrophin releasing factor (CRF) family, and its specific corticotrophin-releasing factor 2 receptor (CRF2R) are highly expressed in skeletal muscle, the role of this peptide in the regulation of skeletal muscle mass and protein metabolism remains elusive. Methods To elucidate the mechanisms how Ucn2 directly controls protein metabolism in skeletal muscles of normal mice, we carried out genetic tools, physiological and molecular analyses of muscles in vivo and in vitro. Results Here, we demonstrated that Ucn2 overexpression activated cAMP signaling and promoted an expressive muscle hypertrophy associated with higher rates of protein synthesis and activation of Akt/mTOR and ERK1/2 signaling pathways. Furthermore, Ucn2 induced a decrease in mRNA levels of atrogin-1 and in autophagic flux inferred by an increase in the protein content of LC3-I, LC3-II and p62. Accordingly, Ucn2 reduced both the transcriptional activity of FoxO in vivo and the overall protein degradation in vitro through an inhibition of lysosomal proteolytic activity. In addition, we demonstrated that Ucn2 induced a fast-to-slow fiber type shift and improved fatigue muscle resistance, an effect that was completely blocked in muscles co-transfected with mitogen-activated protein kinase phosphatase 1 (MKP-1), but not with dominant-negative Akt mutant (Aktmt). Conclusions These data suggest that Ucn2 triggers an anabolic and anti-catabolic response in skeletal muscle of normal mice probably through the activation of cAMP cascade and participation of Akt and ERK1/2 signaling. These findings open new perspectives in the development of therapeutic strategies to cope with the loss of muscle mass.
... Retinol-Binding Protein 4. The protein RBP4 is expressed in adipocytes, myocytes, macrophages, and other tissues and cells (Yang et al., 2005;Broch et al., 2010;Kuperman et al., 2011). The RBP4 facilitates the transport of retinol from the liver to peripheral organs (Berni et al., 1990;Tsutsumi et al., 1992). ...
Article
Full-text available
The involvement of adipose tissue (AT) in metabolism is not limited to energy storage but turned out to be much more complex. We now know that in addition to lipid metabolism, AT is important in glucose homeostasis and AA metabolism and also has a role in inflammatory processes. With the discovery of leptin in 1994, the concept of AT being able to secrete messenger molecules collectively termed as adipokines, and acting in an endo-, para-, and autocrine manner emerged. Moreover, based on its asset of receptors, many stimuli from other tissues reaching AT via the bloodstream can also elicit distinct responses and thus integrate AT as a control element in the regulatory circuits of the whole body's functions. The protein secretome of human differentiated adipocytes was described to comprise more than 400 different proteins. However, in dairy cows, the characterization of the physiological time course of adipokines in AT during the transition from pregnancy to lactation is largely limited to the mRNA level; for the protein level, the analytical methods are limited and available assays often lack sound validation. In addition to proteinaceous adipokines, small compounds such as steroids can also be secreted from AT. Due to the lipophilic nature of steroids, they are stored in AT, but during the past years, AT became also known as being able to metabolize and even to generate ste-roid hormones de novo. In high-yielding dairy cows, AT is substantially mobilized due to increased energy requirements related to lactation. As to whether the steroidogenic system in AT is affected and may change during the common loss of body fat is largely unknown. Moreover, most research about AT in transition dairy cows is based on subcutaneous AT, whereas other depots have scarcely been investigated. This contribution aims to review the changes in adipokine mRNA and where available-protein expression with time relative to calving in high-yielding dairy cows at different conditions, including parity, body condition, diet, specific feed supplements, and health disorders. In addition, the review provides insights into steroidogenic pathways in dairy cows AT and addresses differences between fat depots where possible.
... [14][15][16] UCN2 and CRHR2 play a role in modulating skeletal muscle growth and metabolism. CRHR2 expression is increased in skeletal muscle of high-fat-fed mice, 17 while CRHR2-knockout mice are resistant to high-fat diet (HFD)-induced fat accumulation and insulin resistance. 18 Recent data have shown that UCN3 transgenic mice have increased skeletal muscle mass with myocyte hypertrophy and are protected from obesity. ...
Article
Full-text available
Background Type 2 diabetes and obesity are often seen concurrently with skeletal muscle wasting, leading to further derangements in function and metabolism. Muscle wasting remains an unmet need for metabolic disease, and new approaches are warranted. The neuropeptide urocortin 2 (UCN2) and its receptor corticotropin releasing factor receptor 2 (CRHR2) are highly expressed in skeletal muscle and play a role in regulating energy balance, glucose metabolism, and muscle mass. The aim of this study was to investigate the effects of modified UCN2 peptides as a pharmaceutical therapy to counteract the loss of skeletal muscle mass associated with obesity and casting immobilization. Methods High‐fat‐fed mice (C57Bl/6J; 26 weeks old) and ob/ob mice (11 weeks old) were injected daily with a PEGylated (Compound A) and non‐PEGylated (Compound B) modified human UCN2 at 0.3 mg/kg subcutaneously for 14 days. A separate group of chow‐fed C57Bl/6J mice (12 weeks old) was subjected to hindlimb cast immobilization and, after 1 week, received daily injections with Compound A. In vivo functional tests were performed to measure protein synthesis rates and skeletal muscle function. Ex vivo functional and molecular tests were performed to measure contractile force and signal transduction of catabolic and anabolic pathways in skeletal muscle. Results Skeletal muscles (extensor digitorum longus, soleus, and tibialis anterior) from high‐fat‐fed mice treated with Compound A were ~14% heavier than muscles from vehicle‐treated mice. Chronic treatment with modified UCN2 peptides altered the expression of structural genes and transcription factors in skeletal muscle in high‐fat diet‐induced obesity including down‐regulation of Trim63 and up‐regulation of Nr4a2 and Igf1 (P < 0.05 vs. vehicle). Signal transduction via both catabolic and anabolic pathways was increased in tibialis anterior muscle, with increased phosphorylation of ribosomal protein S6 at Ser235/236, FOXO1 at Ser²⁵⁶, and ULK1 at Ser³¹⁷, suggesting that UCN2 treatment modulates protein synthesis and degradation pathways (P < 0.05 vs. vehicle). Acutely, a single injection of Compound A in drug‐naïve mice had no effect on the rate of protein synthesis in skeletal muscle, as measured via the surface sensing of translation method, while the expression of Nr4a3 and Ppargc1a4 was increased (P < 0.05 vs. vehicle). Compound A treatment prevented the loss of force production from disuse due to casting. Compound B treatment increased time to fatigue during ex vivo contractions of fast‐twitch extensor digitorum longus muscle. Compound A and B treatment increased lean mass and rates of skeletal muscle protein synthesis in ob/ob mice. Conclusions Modified human UCN2 is a pharmacological candidate for the prevention of the loss of skeletal muscle mass associated with obesity and immobilization.
... CRF-R1 and CRF-R2 are found throughout the brain and peripheral tissues (Deussing and Chen, 2018); however, in the brain, CRF-R1 expression is broader than CRF-R2 expression (Van Pett et al., 2000). Conversely, CRF-R2 is broadly expressedrelative to CRF-R1 -in the peripheral tissues, such as skeletal muscle (Kuperman et al., 2011). CRF-R1 exhibits greater affinity for CRF than CRF-R2; UCN2 and UCN3 selectively bind CRF-R2, and UCN1 exhibits equal affinity for both receptors (Deussing and Chen, 2018;Lovejoy and Balment, 1999). ...
Article
The neuroendocrine mechanism underlying stress responses in vertebrates is hypothesized to be highly conserved and evolutionarily ancient. Indeed, elements of this mechanism, from the brain to steroidogenic tissue, are present in all vertebrate groups; yet, evidence of the function and even identity of some elements of the hypothalamus-pituitary-adrenal/interrenal (HPA/I) axis is equivocal among the most basal vertebrates. The purpose of this review is to discuss the functional evolution of the HPA/I axis in vertebrates with a focus on our understanding of this neuroendocrine mechanism in the most ancient vertebrates: the agnathan (i.e., hagfish and lamprey) and chondrichthyan fishes (i.e., sharks, rays, and chimaeras). A review of the current literature presents evidence of a conserved HPA/I axis in jawed vertebrates (i.e., gnathostomes); yet, available data in jawless (i.e., agnathan) and chondrichthyan fishes are limited. Neuroendocrine regulation of corticosteroidogenesis in agnathans and chondrichthyans appears to function through similar pathways as in bony fishes and tetrapods; however, key elements have yet to be identified and the involvement of melanotropins and gonadotropin-releasing hormone in the stress axis in these ancient fishes warrants further investigation. Further, the identities of physiological glucocorticoids are uncertain in hagfishes, chondrichthyans, and even coelacanths. Resolving these and other knowledge gaps in the stress response of ancient fishes will be significant for advancing knowledge of the evolutionary origins of the vertebrate stress response.
... Beside hepatocytes as the main source of RBP4, it is expressed in adipocytes, macrophages, myocytes and further tissues and cells 5,11,12 . Only recently, hepatocytes were identified as the principal source of circulating RBP4 in mice 8 . ...
Article
Full-text available
Retinol binding protein 4 (RBP4) facilitates the transport of retinol in the body but is also an adipokine and fatty acid transporter. Our study was aimed at investigating the associations between RBP4 abundance and fat deposition in cattle. Blood samples of 246 crossbred bulls were taken at 8 months of age and at slaughter at 18 months of age for the determination of RBP4, hormone levels, and fatty acid composition. Significant correlations between plasma RBP4 abundance at 8 months of age and carcass traits at 18 months of age were detected (e.g., r = 0.3; P < 0.001 to carcass fat). Furthermore, RBP4 abundances in the plasma and subcutaneous fat were higher (P < 0.05) in bulls with increased fat deposition, whereas the liver RBP4 expression was not (P > 0.05). Retinol binding protein 4 was immunohistochemically localized in or close to adipocytes within muscle and adipose tissue and in liver stellate cells but not in hepatocytes. Overall, our results indicate that increased RBP4 levels were associated with increased fat deposition and altered fatty acid composition, but not with altered glucose tolerance, in crossbred bulls. Moreover, our results suggest that adipose-tissue-derived RBP4 may contribute to the circulating RBP4 level.
... The CRFR2 is strongly expressed in skeletal muscle and heart (cardiomyocytes), as well as in smooth muscle cells of the vascular system (304,344). In addition, expression has been reported in the pancreas (274). ...
Article
The physiological stress response is responsible for the maintenance of homeostasis in the presence of real or perceived challenges. In this function, the brain activates adaptive responses that involve numerous neural circuits and effector molecules to adapt to the current and future demands. A maladaptive stress response has been linked to the etiology of a variety of disorders, such as anxiety and mood disorders, eating disorders, and the metabolic syndrome. The neuropeptide corticotropin-releasing factor (CRF) and its relatives, the urocortins 1-3, in concert with their receptors (CRFR1, CRFR2), have emerged as central components of the physiological stress response. This central peptidergic system impinges on a broad spectrum of physiological processes that are the basis for successful adaptation and concomitantly integrate autonomic, neuroendocrine, and behavioral stress responses. This review focuses on the physiology of CRF-related peptides and their cognate receptors with the aim of providing a comprehensive up-to-date overview of the field. We describe the major molecular features covering aspects of gene expression and regulation, structural properties, and molecular interactions, as well as mechanisms of signal transduction and their surveillance. In addition, we discuss the large body of published experimental studies focusing on state-of-the-art genetic approaches with high temporal and spatial precision, which collectively aimed to dissect the contribution of CRF-related ligands and receptors to different levels of the stress response. We discuss the controversies in the field and unravel knowledge gaps that might pave the way for future research directions and open up novel opportunities for therapeutic intervention.
... In addition to the central nervous system, CRFR2 is expressed in the periphery, including in the skeletal muscle (10,16,17). Importantly, it has been shown that the levels of CRFR2 in skeletal muscle are elevated by highfat feeding and chronic variable stress (18), conditions that are associated with muscle insulin resistance (19 -24). Moreover, a functional study shows that in primary skeletal muscle cells and in C2C12 myotubes stimulation of CRFR2 by Ucn 2 attenuates phosphorylation of Akt and inhibits insulin-induced glucose uptake (15). ...
Article
Type 2 corticotropin-releasing factor receptor (CRFR2) is expressed in skeletal muscle and stimulation of the receptor has been shown to inhibit the effect of insulin on glucose uptake in muscle cells. Currently, little is known about the mechanisms underlying this process. In this study, we first showed that both in vivo and in vitro CRFR2 expression in muscle was closely correlated with insulin sensitivity, with elevated receptor levels observed in insulin resistant muscle cells. Stimulation of CRFR2 by Urocortin 2 (Ucn 2), a CRFR2 selective ligand, in C2C12 myotubes greatly attenuated insulin-induced glucose uptake. The inhibitory effect of CRFR2 signaling required cAMP production and is involved the mTOR pathway, as rapamycin reversed the inhibitory effect of CRFR2 stimulation on insulin-induced glucose uptake. Moreover, stimulation of CRFR2 failed to inhibit glucose uptake in muscle cells induced by platelet-derived growth factor (PDGF), which, similar to insulin, signals through Akt mediated pathway but is independently of insulin receptor substrate proteins (IRSs) to promote glucose uptake. This result argues that CRFR2 signaling modulates insulin's action likely at the levels of IRS. Consistent with this notion, Ucn 2 reduced insulin-induced tyrosine phosphorylation of IRS-1 and treatment with rapamycin reversed the inhibitory effect of Ucn 2 on IRS-1 and Akt phosphorylation. In conclusion the inhibitory effect of CRFR2 signaling on insulin action is mediated by cAMP in a mTOR dependent manner and IRS-1 is a key nodal point where CRFR2 signaling modulates insulin-stimulated glucose uptake in muscle cells.
Article
The neuropeptide urocortin 2 (UCN2) and its receptor corticotropin-releasing hormone receptor 2 (CRHR2) are highly expressed in skeletal muscle and play a role in regulating energy balance and glucose metabolism. We investigated a modified UCN2 peptide as a potential therapeutic agent for the treatment of obesity and insulin resistance, with a specific focus on skeletal muscle. High-fat-fed mice (C57BL/6J) were injected daily with a PEGylated UCN2 peptide (compound A) at 0.3 mg/kg subcutaneously for 14 days. Compound A reduced body weight, food intake, whole-body fat mass, and intramuscular triglycerides compared with vehicle-treated controls. Furthermore, whole-body glucose tolerance was improved by compound A treatment, with increased insulin-stimulated Akt phosphorylation at Ser473 and Thr308 in skeletal muscle, concomitant with increased glucose transport into extensor digitorum longus and gastrocnemius muscle. Mechanistically, this is linked to a direct effect on skeletal muscle because ex vivo exposure of soleus muscle from chow-fed lean mice to compound A increased glucose transport and insulin signaling. Moreover, exposure of GLUT4-Myc-labeled L6 myoblasts to compound A increased GLUT4 trafficking. Our results demonstrate that modified UCN2 peptides may be efficacious in the treatment of type 2 diabetes by acting as an insulin sensitizer in skeletal muscle.
Article
Full-text available
Two receptors (CRH receptor type 1 and CRH receptor type 2) have been identified for the stress-induced neuropeptide, CRH and related peptides, urocortin, and urocortin II. We previously found marked down-regulation of cardiac CRH receptor type 2 expression following administration of bacterial endotoxin, lipopolysaccharide, a model of systemic immune activation, and inflammation. We postulated that inflammatory cytokines may regulate CRH receptor type 2. We show that systemic IL-1α administration significantly down-regulates CRH receptor type 2 mRNA in mouse heart. In addition, TNFα treatment also reduces CRH receptor type 2 mRNA expression, although the effect was not as marked as with IL-1α. However, CRH receptor type 2 mRNA expression is not altered in adult mouse ventricular cardiomyocytes stimulated in vitro with TNFα or IL-1α. Thus, cytokine regulation may be indirect. Exogenous administration of corticosterone in vivo or acute restraint stress also reduces cardiac CRH receptor type 2 mRNA expression, but like cytokines, in vitro corticosterone treatment does not modulate expression in cardiomyocytes. Interestingly, treatment with urocortin significantly decreases CRH receptor type 2 mRNA in cultured cardiomyocytes. We speculate that in vivo, inflammatory mediators such as lipopolysaccharide and/or cytokines may increase urocortin, which in turn down-regulates CRH receptor type 2 expression in the heart. Because CRH and urocortin increase cardiac contractility and coronary blood flow, impaired CRH receptor type 2 function during systemic inflammation may ultimately diminish the adaptive cardiac response to adverse conditions.
Article
Full-text available
Two receptors (CRH receptor type 1 and CRH receptor type 2) have been identified for the stress-induced neuropeptide, CRH and related peptides, urocortin, and urocortin II. We previously found marked down-regulation of cardiac CRH receptor type 2 expression following administration of bacterial endotoxin, li- popolysaccharide, a model of systemic immune activation, and inflammation. We postulated that inflammatory cytokines may regulate CRH receptor type 2. We show that systemic IL-1a ad- ministration significantly down-regulates CRH receptor type 2 mRNA in mouse heart. In addition, TNFa treatment also reduces CRH receptor type 2 mRNA expression, although the effect was not as marked as with IL-1a. However, CRH receptor type 2 mRNA expression is not altered in adult mouse ventricular car- diomyocytes stimulated in vitro with TNFa or IL-1a. Thus, cy- tokine regulation may be indirect. Exogenous administration of corticosterone in vivo or acute restraint stress also reduces car- diac CRH receptor type 2 mRNA expression, but like cytokines, in vitro corticosterone treatment does not modulate expression in cardiomyocytes. Interestingly, treatment with urocortin sig- nificantly decreases CRH receptor type 2 mRNA in cultured cardiomyocytes. We speculate that in vivo, inflammatory medi- ators such as lipopolysaccharide and/or cytokines may increase urocortin, which in turn down-regulates CRH receptor type 2 expression in the heart. Because CRH and urocortin increase cardiac contractility and coronary blood flow, impaired CRH receptor type 2 function during systemic inflammation may ul- timately diminish the adaptive cardiac response to adverse conditions. (Endocrinology 142: 3537-3545, 2001)
Article
Full-text available
Corticotropin-releasing factor (CRF) and its family of ligands are key regulators of energy balance. These ligands function via activation of their two receptors, CRFR1 and CRFR2. CRFR1 has been shown to be the dominant receptor in acti- vation of the hypothalamic-pituitary-adrenal axis in response to stress as well as a key mediator of anxiety in the limbic system. To specifically examine the role of CRFR2 in energy balance, mice deficient for CRFR2 were exposed to physio- logical perturbations of homeostasis, including high-fat diet, repeated cold stress, and glucose and insulin challenges, and their responses measured. While on a high-fat diet, CRFR2- mutant mice consumed substantially more food and main- taining the same weight but had significantly lower body fat and lower plasma lipids than their wild-type littermates. These mice were also less inclined to develop diet-induced insulin resistance and more sensitive to changes in plasma glucose, indicating increased insulin sensitivity. Following repeated cold stress, mutant mice had significantly lower body fat and a transient reduction in feed efficiency, despite similar body weights, suggesting a possible preference for fat as an energy substrate. Elevated levels of uncoupling pro- tein-1 in brown adipose tissue as well as smaller white and brown adipocytes from CRFR2-mutant mice were indications of possible increased sympathetic tone. These results demon- strate that CRFR2 plays a critical role in regulation of energy expenditure and is important for responses to homeostatic challenges. (Endocrinology 144: 2580 -2587, 2003)
Article
Full-text available
In obesity and type 2 diabetes, expression of the GLUT4 glucose transporter is decreased selectively in adipocytes. Adipose-specific Glut4 (also known as Slc2a4) knockout (adipose-Glut4 -/-) mice show insulin resistance secondarily in muscle and liver. Here we show, using DNA arrays, that expression of retinol binding protein-4 (RBP4) is elevated in adipose tissue of adipose-Glut4 -/- mice. We show that serum RBP4 levels are elevated in insulin-resistant mice and humans with obesity and type 2 diabetes. RBP4 levels are normalized by rosiglitazone, an insulin-sensitizing drug. Transgenic overexpression of human RBP4 or injection of recombinant RBP4 in normal mice causes insulin resistance. Conversely, genetic deletion of Rbp4 enhances insulin sensitivity. Fenretinide, a synthetic retinoid that increases urinary excretion of RBP4, normalizes serum RBP4 levels and improves insulin resistance and glucose intolerance in mice with obesity induced by a high-fat diet. Increasing serum RBP4 induces hepatic expression of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) and impairs insulin signalling in muscle. Thus, RBP4 is an adipocyte-derived 'signal' that may contribute to the pathogenesis of type 2 diabetes. Lowering RBP4 could be a new strategy for treating type 2 diabetes.
Article
Corticotropin-releasing hormone (CRH) is the principal regulator of the stress response. CRH stimulates production of ACTH via specific CRH receptors located on pituitary corticotropes. In addition to pituitary and central nervous system effects, peripheral effects of CRH have been observed involving the immune and cardiovascular systems. Specific CRH binding studies in several peripheral organs, as well as functional studies, have implied the existence of peripheral CRH receptors. Although a pituitary/brain CRH receptor has recently been identified, it is expressed at very low levels in peripheral sites where CRH effects have been observed. We report here the identification of a novel murine CRH receptor that is highly expressed in the heart. The newly cloned CRH receptor cDNA (CRH-R2) was isolated from a mouse heart cDNA library and encodes a 430-amino acid protein containing seven putative transmembrane domains characteristic of G protein-coupled receptors. CRH-R2 is 69% identical with the previously i...
Article
CRH induces corticotrope membrane depolarization and facilitates action potential firing. The increase in electrical excitability causes large oscillatory increases in cytosolic Ca”- levels. In this study on highly enriched populations of cultured rat corticotropes, inhibitors were used to determine the contribution ofthe Na’ channel and Ca” channel subtypes to membrane excitability and cytosolic Ca”+ levels. Tetrodotoxin, an inhibitor of the voltage-dependent Na’ channel, inhibited a rapid initial component of the action potential, but generally did not influence spontaneous or CRH-induced firing frequency. Tetrodotoxin also had no effect on spontaneous or CRHinduced cytosolic Ca*’ levels. The L-type Ca“ channel inhibitor nifedipine abolished spontaneous and CRH-induced action potentials and cytosolic Ca’ + transients, but did not eliminate the CRH-induced membrane depolarization or completely restore cytosolic Ca”’ to basal levels. Inhibition of P-type Ca”’ channels with w-agatoxin-IVA decreased action potential firing frequency and reduced the CRHinduced increase in cytosolic Ca”. The combination of nifedipine and w-agatoxin-IVA abolished the CRH-induced rise in Ca2- but did not abolish the membrane depolarization. Thus, cytosolic Ca’ + is mainly increased by CRH-induced action potentials that are completely dependent on L-type Ca” channels and partially regulated by P-type Ca”- channels. CRH-induced Ca” entry also occurs independently of action potentials and is due to P-type, and possibly L-type, Ca”’ channels activated by the CRH-induced membrane depolarization.
Article
The product of the ob gene, leptin, is thought to act in the hypothalamus to reduce food intake and body weight (b.w.) in rats and mice; however, the mechanisms of leptin action in the brain have not been fully elucidated. Corticotropin-releasing hormone (CRH) is a potent anorectic neuropeptide, and its type-2 receptor (CRHR-2) in the ventromedial hypothalamus (VMH) appears to play an important role in the expression of this anorectic effect. We explored here the impact of systemic leptin administration on CRH mRNA expression in the hypothalamic paraventricular nucleus (PVN) and CRHR-2 mRNA expression in the VMH in male rats, using in-situ hybridization histochemistry. The expression of CRH mRNA in the PVN and CRHR-2 mRNA in the VMH were increased at 2 h and 6 h, respectively, after a single intraperitoneal injection of leptin (1.0 mg/kg). Continuous subcutaneous infusion of leptin (1.2 mg/kg/day) via an osmotic minipump for 5 days increased the expression of CRHR-2 mRNA in the VMH, but not the expression of CRH mRNA in the PVN, compared with vehicle treatment. The rats that received the single or continuous administration of leptin showed reductions of food intake and b.w. compared with vehicle-treated rats. These results are consistent with our previous findings that the expression of CRHR-2 mRNA in the VMH is positively correlated with plasma leptin concentrations under various conditions, and highlight the importance of circulating leptin for the regulation of VMH CRHR-2 mRNA. The present results also raise the possibility that leptin reduces food intake and b.w. at least partially due to the enhancement of the anorectic effect of CRH via increased PVN CRH expression and/or VMH CRHR-2 expression.