ArticlePDF Available

Calcineurin Mediates the Calcium-dependent Inhibition of Adipocyte Differentiation in 3T3-L1 Cells

Authors:

Abstract and Figures

Recent studies have revealed that the calcium-dependent serine/threonine phosphatase calcineurin mediates the effects of intracellular calcium in many different cell types. In this study we investigated the role of calcineurin in the regulation of adipocyte differentiation. We found that the specific calcineurin inhibitors cyclosporin A and FK506 overcame the antiadipogenic effect of calcium ionophore on the differentiation of 3T3-L1 preadipocytes. This finding suggests that calcineurin is responsible for mediating the previously documented Ca(2+)-dependent inhibition of adipogenesis. We further demonstrate that the expression of a constitutively active calcineurin mutant potently inhibits the ability of 3T3-L1 cells to undergo adipocyte differentiation by preventing expression of the proadipogenic transcription factors peroxisome proliferator-activated receptor gamma (PPARgamma) and CCAAT/enhancer-binding protein alpha (C/EBPalpha). This calcineurin-mediated block in adipocyte differentiation is rescued by ectopic expression of PPARgamma1. Finally, we demonstrate that inhibition of endogenous calcineurin activity with either FK506 or a specific calcineurin inhibitory peptide enhances differentiation of 3T3-L1 cells in response to suboptimal adipogenic stimuli, suggesting that endogenous calcineurin activity normally sets a signaling threshold that antagonizes efficient adipocyte differentiation. Collectively, these data indicate that calcineurin acts as a Ca(2+)-dependent molecular switch that negatively regulates commitment to adipocyte differentiation by preventing the expression of critical proadipogenic transcription factors.
Content may be subject to copyright.
Calcineurin Mediates the Calcium-dependent Inhibition of
Adipocyte Differentiation in 3T3-L1 Cells*
Received for publication, August 2, 2002, and in revised form, September 22, 2002
Published, JBC Papers in Press, September 25, 2002, DOI 10.1074/jbc.M207913200
Joel W. Neal and Neil A. Clipstone‡
From the Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University,
Chicago, Illinois 60611
Recent studies have revealed that the calcium-
dependent serine/threonine phosphatase calcineurin
mediates the effects of intracellular calcium in many
different cell types. In this study we investigated the
role of calcineurin in the regulation of adipocyte differ-
entiation. We found that the specific calcineurin inhib-
itors cyclosporin A and FK506 overcame the antiadipo-
genic effect of calcium ionophore on the differentiation
of 3T3-L1 preadipocytes. This finding suggests that cal-
cineurin is responsible for mediating the previously
documented Ca
2
-dependent inhibition of adipogenesis.
We further demonstrate that the expression of a consti-
tutively active calcineurin mutant potently inhibits the
ability of 3T3-L1 cells to undergo adipocyte differentia-
tion by preventing expression of the proadipogenic
transcription factors peroxisome proliferator-activated
receptor
(PPAR
) and CCAAT/enhancer-binding pro-
tein
(C/EBP
). This calcineurin-mediated block in adi-
pocyte differentiation is rescued by ectopic expression
of PPAR
1. Finally, we demonstrate that inhibition of
endogenous calcineurin activity with either FK506 or a
specific calcineurin inhibitory peptide enhances differ-
entiation of 3T3-L1 cells in response to suboptimal adi-
pogenic stimuli, suggesting that endogenous cal-
cineurin activity normally sets a signaling threshold
that antagonizes efficient adipocyte differentiation. Col-
lectively, these data indicate that calcineurin acts as a
Ca
2
-dependent molecular switch that negatively regu-
lates commitment to adipocyte differentiation by pre-
venting the expression of critical proadipogenic tran-
scription factors.
Adipocytes are highly specialized cells that play a key role in
energy homeostasis by regulating the storage and release of
energy in response to changing nutritional needs (1). In addi-
tion to their role in energy balance, adipocytes also perform
important endocrine functions by secreting a variety of factors
that regulate such processes as food intake, insulin responsive-
ness, reproduction, vascular remodeling, and the immune re-
sponse (2). Although adipocytes clearly play an important
physiological role, the excessive accumulation of adipose tissue
can result in obesity, which is known to be a significant risk
factor for a number of other disease states including insulin
resistance, type-2 diabetes, hypertensions, cardiovascular dis-
ease and cancer (3). Obesity can arise from either an increase
in individual adipocyte cell size or from an increase in total
adipocyte cell number as a result of increased de novo adipocyte
differentiation (1). Accordingly, the molecular mechanisms
that govern the regulation of adipocyte growth and differenti-
ation are of considerable scientific interest and have been the
subject of much investigation (1, 4, 5).
Considerable progress in our understanding of adipocyte bi-
ology has come from the study of the 3T3-L1 preadipocyte cell
line (6), which under the appropriate in vitro culture conditions
can be efficiently induced to undergo terminal differentiation
into morphologically distinct, triglyceride-laden, mature adipo-
cytes. Adipocyte differentiation is induced in 3T3-L1 preadipo-
cytes by treatment of confluent, growth-arrested cells with the
adipogenic hormones methylisobutylxanthine (Mix),
1
dexam-
ethasone (Dex), and insulin, collectively known as MDI. A large
body of accumulated data has revealed that this process of
adipocyte differentiation proceeds via a highly orchestrated
and coordinated cascade of transcription factors, including
members of the CCAAT/enhancer-binding protein (C/EBP)
family and peroxisome proliferator-activated receptor
(PPAR
) (1, 4, 5). MDI-treated, growth-arrested 3T3-L1 prea-
dipocytes synchronously enter the cell cycle and initially ex-
press the early transcription factors C/EBP
and C/EBP
(7, 8).
C/EBP
and C/EBP
then elicit the expression of the proadi-
pogenic transcription factor PPAR
(7), which in turn induces
the expression of C/EBP
(9). Together, PPAR
and C/EBP
then are believed to play a dual role in adipogenesis by first
inducing withdrawal from the cell cycle and then directing the
expression of adipocyte-specific genes that ultimately result in
the acquisition of the mature adipocyte cell fate (10 –13).
This process of adipocyte differentiation is influenced by a
variety of different extrinsic factors and intracellular signaling
pathways (4, 14). Of particular interest to the current study are
the effects of intracellular calcium on adipocyte differentiation.
A number of reports have demonstrated that increases in in-
tracellular calcium concentration ([Ca
2
]
i
) during the early
phase of human and 3T3-L1 preadipocyte differentiation act to
potently inhibit adipogenesis (15–17). However, the effects of
calcium on this process may be complex, because increases in
[Ca
2
]
i
in human preadipocytes during the later stages of dif-
* This work was supported in part by a Gramm travel fellowship
award from the Robert H. Lurie Comprehensive Cancer Center of
Northwestern University (to J. W. N.) and by National Institutes of
Health Grant R29 GM55292 (to N. A. C.). The costs of publication of
this article were defrayed in part by the payment of page charges. This
article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
To whom correspondence should be addressed: Dept. of Microbiol-
ogy-Immunology, Northwestern University, 303 E. Chicago Ave., Chi-
cago, IL 60611. Tel.: 312-503-8233; Fax: 312-503-1339; E-mail:
n-clipstone@northwestern.edu.
1
The abbreviations used are: Mix, methylisobutylxanthine; Dex, dex-
amethasone; MDI, methylisobutylxanthine, dexamethasone and insu-
lin; C/EBP, CCAAT/enhancer-binding protein; PPAR
, peroxisome pro-
liferator activated receptor
; NFAT, nuclear factor of activated T cells;
[Ca
2
]
i
, intracellular calcium concentration; CsA, cyclosporin A;
CNmut, constitutively activated calcineurin mutant; MSCV, murine
stem cell virus; IRES, internal ribosomal entry sequence; pEGFP, per-
muted enhanced green fluorescent protein; Ab, antibody; LTR, long
terminal repeat.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 277, No. 51, Issue of December 20, pp. 49776–49781, 2002
© 2002 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org49776
by guest on November 6, 2015http://www.jbc.org/Downloaded from
ferentiation and in Ob1774 cells appear to enhance the expres-
sion of certain markers of mature adipocytes (1719). In this
study we investigated the molecular mechanism that underlies
the inhibitory effect of early increases in [Ca
2
]
i
on the differ-
entiation of 3T3-L1 preadipocytes. Calcineurin, a calcium-de-
pendent serine/threonine phosphatase, is known to be a critical
downstream effector of the calcium signal in a wide variety of
different cell types (20). We present evidence to demonstrate
that calcineurin mediates the Ca
2
-dependent inhibition of
adipocyte differentiation in 3T3-L1 cells. In addition, we dem-
onstrate that inhibition of endogenous calcineurin activity in
3T3-L1 cells enhances the efficiency of adipogenesis in re-
sponse to suboptimal adipogenic stimuli. On the basis of our
results, we propose that commitment to the terminal phase of
adipocyte differentiation is likely to be regulated by the level of
calcineurin activity in preadipocytes.
EXPERIMENTAL PROCEDURES
Cell Culture and Adipocyte Differentiation—3T3-L1 preadipocytes
(ATCC) were cultured in a growth medium of Dulbeccos modified
Eagles medium with high glucose (Invitrogen) supplemented with 10%
(v/v) fetal calf serum (Hyclone), 100 units/ml penicillin G, and 100
g/ml
streptomycin (Invitrogen). To induce adipocyte differentiation, cells
were grown until 2 days postconfluence (day 0) and then treated for 2
days with growth medium plus MDI (0.5 mMmethylisobutylxanthine, 1
Mdexamethasone, and 10
g/ml insulin, all from Sigma). The cells
were re-fed with growth medium that contained 10
g/ml insulin at day
2 and every 2 days thereafter with growth medium alone. After 10 days,
cells were fixed with formalin and stained with the lipophilic dye Oil
Red O (Sigma). Stained cells were either photographed or counter-
stained with Giemsa and visualized by bright field microscopy. Where
indicated, cells were treated additionally with 2
Mionomycin, 5 ng/ml
FK506, 1
g/ml CsA (all from Calbiochem), or vehicle control (ethanol).
Retroviral Expression Constructs—The retroviral expression vector
pMSCV-CNmut was generated by insertion of a XhoI-EcoRI fragment
that contained the previously described CNmut (21) into pMSCV-GFP
downstream of the viral long terminal repeat and upstream of the
IRES-GFP cassette. The pMSCV-H2K retroviral expression vector was
created by replacing GFP in the pMSCV-GFP retroviral expression
vector with a PCR-amplified truncated murine major histocompatibility
class I H-2K
k
cDNA from pMACS Kk.II (Miltenyi Biotec). pMSCV-
PPAR
1 was created by introducing the full-length, cDNA-encoding
murine PPAR
1 (a gift from J. Reddy, Northwestern University) into
pMSCV-H2K. pMSCV-VIVIT-GFP was constructed by inserting an ol-
igonucleotide that encoded a previously described (22) calcineurin-in-
hibitory peptide (MAGPHPVIVITGPHEE) into pEGFP-N3 (Clontech)
and then introducing the resulting VIVIT-GFP fusion sequence into
pMSCV-H2K.
Retrovirus Production and Infection of 3T3-L1 Cells—Retroviral ex-
pression vectors were cotransfected with pVSV-G (Clontech) into the
GP293 pantropic packaging cell line (Clontech) by using Lipo-
fectAMINE Plus (Invitrogen). The medium was replaced after 24 h, and
viral supernatants were harvested at 2 days post-transfection and
stored at 80 °C. For infections, 5 10
4
3T3-L1 cells were plated per
well of a 6-well plate. The next day, the medium was replaced with 2 ml
of viral supernatant that contained 8
g/ml polybrene (Sigma), and
plates were centrifuged at 2000 rpm for 1.5 h at room temperature.
After removal of the viral supernatant, cells were expanded in growth
medium for subsequent analysis. For double infections, previously in-
fected cells were replated after 3 days and infected with the second
virus as described previously.
Flow Cytometric Analysis—On day 3 after infection, cells were ana-
lyzed for GFP fluorescence or were stained additionally with a phyco-
erythrin-coupled anti-mouse H-2K
k
Ab (36-7-5; BD Biosciences). 10,000
events were analyzed using a FACSCaliber flow cytometer and
CELLQuest software (BD Biosciences).
Immunoblot and Northern Blot Analysis—Protein extracts prepared
from cells harvested at the indicated times postdifferentiation were
resolved by SDS-PAGE and subjected to immunoblot analysis with the
relevant Ab. All Abs (PPAR
(H-100), C/EBP
(14AA), C/EBP
(H-7),
and C/EBP
(C-22)) were purchased from Santa Cruz Biotechnology.
For Northern blot analysis, total RNA was isolated from cells by using
Trizol (Invitrogen) on the indicated day after differentiation was in-
duced. RNA samples (10
g) were separated by using 1.2% agarose, 2.2
Mformaldehyde gel electrophoresis and transferred to Hybond-N mem-
brane (Amersham Biosciences). Immobilized RNA was hybridized with
a
32
P-radiolabeled murine aP2 probe cDNA probe (ATCC) and visual-
ized by exposure to Kodak X-AR film. Membranes were stripped and
reprobed with a glyceraldehyde-3-phosphate dehydrogenase probe as a
control.
RESULTS
Calcineurin Is Required for the Ca
2
-dependent Inhibition of
Adipocyte Differentiation—To investigate the role of cal-
cineurin in the regulation of adipocyte differentiation, we first
examined whether the specific calcineurin inhibitors FK506
and CsA were able to attenuate the previously reported inhib-
itory effect of calcium ionophore on the differentiation of the
3T3-L1 preadipocyte cell line (15). As shown in Fig. 1, 2-day
postconfluent plates of 3T3-L1 cells that were treated with
MDI efficiently differentiated into morphologically distinct, fat-
laden adipocytes with accumulated cytoplasmic triglycerides
that stained red with Oil Red O. Notably, the presence of either
FK506 or CsA did not affect the ability of MDI to induce 3T3-L1
cells to undergo adipocyte differentiation. This fact indicates
that calcineurin is not required for MDI-induced adipocyte
differentiation, as was originally proposed by Ho et al. (23).
Consistent with a previous report (15), treatment of 3T3-L1
cells with the calcium ionophore ionomycin potently blocked
their differentiation. Significantly, we found that this inhibi-
tory effect of calcium ionophore was abrogated in the presence
of either FK506 or CsA (Fig. 1). These data therefore indicate
that calcineurin activity is required to mediate the inhibitory
effects of calcium ionophore on the differentiation of 3T3-L1
preadipocytes into mature adipocytes and suggest that cal-
cineurin is likely to negatively regulate adipocyte
differentiation.
A Calcium-independent, Constitutively Active Form of Cal-
cineurin Inhibits Adipocyte Differentiation in 3T3-L1 Cells—To
FIG.1. The immunosuppressive drugs CsA and FK506 over-
come the antiadipogenic effects of the calcium ionophore, iono-
mycin. 3T3-L1 preadipocytes were induced to undergo adipocyte dif-
ferentiation by treatment with MDI as described under Experimental
Procedures.As indicated, cells were additionally treated for the first 4
days of differentiation in the presence of 2
Mionomycin or vehicle
(ethanol), plus either vehicle (ethanol), 5 ng/ml FK506, or 1
g/ml CsA.
After 10 days, plates of cells were fixed, stained with Oil Red O, and
either directly photographed or counterstained with Giemsa and visu-
alized by bright field microscopy.
Calcineurin Negatively Regulates Adipocyte Differentiation 49777
by guest on November 6, 2015http://www.jbc.org/Downloaded from
further investigate the effects of calcineurin on adipogenesis,
we used an efficient retroviral gene delivery system to intro-
duce a previously characterized (21) calcium-independent, con-
stitutively active calcineurin mutant (CNmut) into 3T3-L1
cells. The cDNA that encoded CNmut was introduced into the
MSCV-GFP retroviral vector under the control of the MSCV
promoter and upstream of an IRES-GFP expression cassette,
thereby allowing the expression of both CNmut and GFP from
a single bicistronic mRNA (Fig. 2A). Using these vectors, we
were routinely able to generate a high-titer retrovirus capable
of stably infecting 95% of 3T3-L1 cells (Fig. 2B). As shown in
Fig. 2C, 3T3-L1 preadipocytes infected with the control MSCV-
GFP retrovirus and treated with MDI efficiently differentiated
into mature Oil Red O-staining adipocytes. In contrast, we
found that cells infected with the MSCV-CNmut retrovirus and
stimulated with MDI failed to undergo the characteristic mor-
phological changes associated with adipocyte differentiation
and did not stain red with Oil Red O. As expected, we were able
to rescue adipogenesis in these CNmut-expressing cells by
treatment with FK506 (data not shown). To confirm the inhib-
itory effect of CNmut on adipocyte differentiation, we next
examined the time course of expression of the late adipocyte-
specific marker gene, aP2. As shown in Fig. 2D, expression of
aP2 mRNA was readily detectable in control MSCV-GFP-in-
fected cells after treatment with MDI, whereas this transcript
was not detectable in MDI-induced CNmut-expressing cells.
Taken together, these results indicate that sustained cal-
cineurin activity in 3T3-L1 preadipocytes inhibits adipocyte
differentiation.
Sustained Calcineurin Activity Inhibits the Expression of the
Proadipogenic Transcription Factors PPAR
and C⁄EBP
The transcription factors PPAR
and C/EBP
are known to be
both necessary and sufficient for adipocyte differentiation (10,
11, 24, 25). To investigate the molecular mechanism that un-
derlies the inhibitory effect of calcineurin on adipocyte differ-
entiation, we next examined the effects of CNmut on the ex-
pression of PPAR
and C/EBP
. Thus, cell extracts prepared
from 3T3-L1 cells infected with either MSCV-GFP or MSCV-
CNmut that had been induced to undergo adipocyte differenti-
ation by treatment with MDI were analyzed for expression of
PPAR
and C/EBP
by immunoblot analysis. As shown in Fig.
3A, the expression of both PPAR
and C/EBP
was readily
detectable in cells infected with MSCV-GFP. In contrast, we
did not observe any appreciable expression of either PPAR
or
C/EBP
in CNmut-expressing cells (Fig. 3A). Thus, calcineurin
activity appears to inhibit adipocyte differentiation by prevent-
ing the expression of the proadipogenic transcription factors
PPAR
and C/EBP
.
Sustained Calcineurin Activity Does Not Affect the Induction
of C⁄EBP
and C⁄EBP
The expression of PPAR
and
C/EBP
is thought to be regulated during adipocyte differen-
tiation by the transcription factors C/EBP
and C/EBP
(7, 8).
We therefore examined whether calcineurin activity blocks the
induction of PPAR
and C/EBP
by interfering with the ex-
pression of C/EBP
and C/EBP
. As shown in Fig. 3B, exposure
of MSCV-GFP-infected control cells to differentiation-inducing
conditions resulted in the expression of both C/EBP
and
FIG.2. A calcium-independent, constitutively active form of
calcineurin inhibits adipocyte differentiation in 3T3-L1 cells. A,
schematic representation of the MSCV-GFP and MSCV-CNmut retro-
viral vectors. B, flow cytometric analysis of GFP expression after ret-
roviral infection of 3T3-L1 preadipocytes with the MSCV-GFP and
MSCV-CNmut retroviruses. The percentage of GFP expressing 3T3-L1
cells (green) as compared with mock infected cells (gray) is indicated. C,
3T3-L1 preadipocytes infected with either MSCV-GFP or MSCV-
CNmut retroviruses were induced to undergo differentiation as de-
scribed under Experimental Procedures.After 10 days, cells were
stained with Oil Red O. D, Northern blot analysis of aP2 mRNA ex-
pression in MSCV-GFP- and MSCV-CNmut-infected cells induced to
undergo differentiation for the indicated number of days (upper panel).
The membrane was reprobed by using glyceraldehyde-3-phosphate de-
hydrogenase (GAPDH) as a loading control (lower panel).
FIG.3.Effects of sustained calcineurin activity on the expres-
sion of adipogenic transcription factors. 3T3-L1 preadipocytes
infected with either MSCV-GFP or MSCV-CNmut retroviruses were
induced to undergo differentiation with MDI as described under Ex-
perimental Procedures.Whole cell extracts were prepared at the indi-
cated time points and analyzed by SDS-PAGE followed by immunoblot-
ting with the indicated Ab: PPAR
(A,upper panel), C/EBP
(A,lower
panel), C/EBP
(B,upper panel), C/EBP
(B,lower panel).
Calcineurin Negatively Regulates Adipocyte Differentiation49778
by guest on November 6, 2015http://www.jbc.org/Downloaded from
C/EBP
. We found that ectopic expression of CNmut did not
attenuate the expression of C/EBP
and C/EBP
(unlike
PPAR
and C/EBP
), because both were induced in CNmut-
expressing cells with similar kinetics to control cells (Fig. 3B).
Moreover, it appeared that calcineurin did not affect the rela-
tive abundance of the smaller inhibitory liver-enriched inhibi-
tory protein (LIP) isoform of C/EBP
that arises from alterna-
tive translational initiation and is believed to represent a
naturally occurring dominant-negative regulator of C/EBP
family members (26). Thus, it appears that sustained cal-
cineurin activity does not prevent the induction of PPAR
and
C/EBP
by affecting the expression of the early transcription
factors C/EBP
and C/EBP
.
Ectopic Expression of PPAR
1 Rescues Adipocyte Differenti-
ation in CNmut-expressing 3T3-L1 CellsWe wanted to deter-
mine next whether the inhibitory effect of CNmut on adipocyte
differentiation was specifically caused by prevention of the
expression of PPAR
and C/EBP
, or whether sustained cal-
cineurin activity might merely nonspecifically perturb 3T3-L1
cellular physiology by creating a cellular environment incom-
patible with cellular differentiation. To distinguish between
these possibilities, we tested whether ectopic expression of
PPAR
was able to bypass the block in adipogenesis and rescue
adipocyte differentiation in CNmut-expressing cells. Thus,
3T3-L1 cells were infected sequentially first with MSCV-
CNmut and then with either MSCV-PPAR
1 or MSCV-H2K as
a control. The MSCV-PPAR
1 retroviral vector directs the ex-
pression of both PPAR
1 and the murine major histocompati-
bility class I molecule, H2K
k
from a single bicistronic mRNA
(Fig. 4A). As a result, successful infection with this virus can be
monitored readily by fluorescence-activated cell sorter analysis
with a fluorescence-conjugated anti-H2K
k
mAb. Using this se-
quential infection protocol, we were able to doubly infect 95%
of cells with MSCV-CNmut and either MSCV-H2K or MSCV-
PPAR
1 (Fig. 4B). As expected, cells infected with both MSCV-
CNmut and MSCV-H2K failed to undergo adipocyte differen-
tiation under standard differentiation conditions (Fig. 4C). In
contrast, cells that co-expressed both CNmut and PPAR
1
were found to efficiently undergo the characteristic morpholog-
ical changes associated with adipocyte differentiation and
stained positive for Oil Red O (Fig. 4C). Fluorescent micro-
scopic analysis of these cells revealed that they still expressed
the CNmut-IRES-GFP transgene, ruling out the trivial possi-
bility that PPAR
1 rescued adipogenesis by inhibiting expres-
sion of CNmut (data not shown). Furthermore, we observed
that expression of PPAR
1 overcomes the inhibitory effects of
ionomycin on adipogenesis (Fig. 4D). The efficient rescue of
adipocyte differentiation in both CNmut-expressing and iono-
mycin-treated cells by ectopic expression of PPAR
1 suggests
that calcineurin principally inhibits adipogenesis by prevent-
ing the expression of the proadipogenic transcription factor
PPAR
.
Inhibition of Endogenous Calcineurin Activity Enhances Adi-
pocyte DifferentiationHaving demonstrated that the sus-
tained activation of calcineurin either by treatment with cal-
cium ionophore or ectopic expression of CNmut potently
inhibits adipogenesis, we wanted to investigate the potential
role of calcineurin during the normal process of in vitro adipo-
cyte differentiation. For these experiments, we took advantage
of our observation that treatment of 3T3-L1 preadipocytes with
suboptimal adipogenic stimuli (Mix and Dex without insulin)
resulted in only modest adipocyte differentiation that occurred
primarily in isolated patches of cells. As seen in Fig. 5A, stim-
ulation of 3T3-L1 cells with decreasing concentrations of Mix
and Dex resulted in a dose-dependent decrease in adipocyte
differentiation. However, when 3T3-L1 cells were differenti-
ated under these suboptimal conditions in the presence of ei-
ther FK506 or CsA, we observed enhanced differentiation with
a significant increase in both the number and the size of these
adipogenic cell clusters (Fig. 5Aand data not shown). To fur-
ther implicate endogenous calcineurin in the regulation of adi-
pocyte differentiation, we took advantage of a previously char-
acterized specific peptide inhibitor of calcineurin, VIVIT-GFP,
which has been shown to specifically inhibit the ability of
calcineurin to activate NFAT proteins (22). As predicted, ex-
pression of VIVIT-GFP in 3T3-L1 cells was able to overcome
the inhibitory effects of ionomycin on adipocyte differentiation
(Fig. 5B). Consistent with the effect of FK506 (Fig. 5A), we
found that inhibition of endogenous calcineurin activity with
VIVIT-GFP dramatically enhanced adipocyte differentiation in
response to suboptimal adipogenic stimuli (Fig. 5C). Taken
together, these results demonstrate that endogenous cal-
cineurin activity acts to antagonize the normal process of adi-
pogenesis and is likely to set a signaling threshold required for
efficient adipocyte differentiation.
DISCUSSION
In the current study, we provide multiple lines of evidence to
indicate that the Ca
2
-calmodulin-regulated phosphatase cal-
cineurin acts to negatively regulate adipocyte differentiation.
We demonstrate that the activation of the calcineurin signaling
pathway inhibits adipogenesis by preventing the expression of
the proadipogenic transcription factors PPAR
and C/EBP
.
FIG.4.Ectopic expression of PPAR
rescues adipocyte differ-
entiation in CNmut-expressing 3T3-L1 cells. A, schematic repre-
sentation of the MSCV-PPAR
retroviral vector. LTR, long terminal
repeat. B, representative flow cytometric analysis of GFP and H2K
k
expression after double infection of 3T3-L1 preadipocytes with the
MSCV-CNmut and MSCV-H2K retroviruses. Data are presented as
two-color dot plots, and the percentage of cells infected with both
retroviruses is indicated. C, 3T3-L1 preadipocytes infected with MSCV-
CNmut and either MSCV-PPAR
or control MSCV-H2K virus were
induced to undergo differentiation as described under Experimental
Procedures.After 10 days, cells were stained with Oil Red O. D, 3T3-L1
preadipocytes were infected with either MSCV-H2K or MSCV-PPAR
and then induced to undergo differentiation with MDI in the presence
of ionomycin as described under Experimental Procedures.
Calcineurin Negatively Regulates Adipocyte Differentiation 49779
by guest on November 6, 2015http://www.jbc.org/Downloaded from
Conversely, we found that inhibition of endogenous calcineurin
activity markedly enhances adipocyte differentiation in 3T3-L1
cells in response to suboptimal adipogenic stimuli. Together
our findings suggest that the level of activation of the endoge-
nous calcineurin signaling pathway in preadipocyte cells is
likely to play an important role in setting the signaling thresh-
old required for commitment to the terminal phase of adipocyte
differentiation. Given that calcineurin activity is exquisitely
sensitive to changes in [Ca
2
]
i
(27), the role of calcineurin in
the regulation of adipocyte differentiation is likely to be espe-
cially important in the response to environmental cues such as
prostaglandin F
2
that modify the level of intracellular Ca
2
.
Prostaglandin F
2
has been shown previously to inhibit adipo-
genesis by a Ca
2
-dependent mechanism (16). On the basis of
our findings, we propose that calcineurin is an intrinsic nega-
tive regulatory component of the adipogenic signaling pathway
that acts as a Ca
2
-dependent molecular switch to inhibit
adipocyte differentiation in response to exogenous agents that
elevate [Ca
2
]
i
.
It is important to note that our primary conclusion that
calcineurin acts to negatively regulate adipocyte differentiation
conflicts with a previous study by Ho et al. (23). On the basis of
their identification of an NFAT binding site in a proximal
promoter element of the adipocyte-specific aP2 gene and their
observation that CsA inhibits MDI-induced 3T3-L1 differenti-
ation, these authors proposed a positive role for the calcineurin/
NFAT signaling pathway in the regulation of adipogenesis.
However, a previous study by Yeh et al. (28) found that neither
CsA nor FK506 inhibited the differentiation of 3T3-L1 cells. In
fact, they found that a molar excess of FK506 was able to
overcome the antiadipogenic effects of the structurally related
drug rapamycin. Because both FK506 and rapamycin are
known to mediate their biological activities by binding to a
common intracellular receptor, FK506-binding protein (29), the
ability of FK506 to overcome the antiadipogenic effects of ra-
pamycin indicates that functional FK506-FK506 binding pro-
tein complexes are unable to block adipogenesis. This observa-
tion and our current data argue strongly against a positive role
for calcineurin in adipocyte differentiation, as originally sug-
gested by Ho et al. (23). Instead, our data provide multiple
independent lines of evidence that demonstrate a novel inhib-
itory function for calcineurin in the regulation of adipocyte
differentiation. At present, the reason behind the discrepancy
between our data and those of Ho et al. is not clear but may be
related to either the method of drug delivery or specific cell
culture conditions.
Our finding that sustained calcineurin activity inhibits adi-
pocyte differentiation by preventing the expression of PPAR
and C/EBP
, but not C/EBP
and C/EBP
, suggests a number
of potential mechanisms by which calcineurin may inhibit ad-
ipogenesis. First, calcineurin may interfere directly with the
activity of C/EBP
and C/EBP
. Although C/EBP
is thought
to be regulated during 3T3-L1 differentiation by a phosphoryl-
ation-dependent mechanism (30), no evidence currently exists
to suggest that C/EBP
is a direct substrate of calcineurin. In
addition, it appears that calcineurin does not affect the expres-
sion of known inhibitors of C/EBP
activity such as LIP (Fig.
3B), an inhibitory C/EBP
isoform that arises by alternative
translational initiation (26), or the expression of CHOP-10
(data not shown), which is believed to represent an endogenous
dominant-negative inhibitor of the C/EBP family (31). Second,
calcineurin may inhibit adipocyte differentiation by affecting a
parallel pathway to C/EBP
and C/EBP
that is also required
for the efficient expression of PPAR
and C/EBP
. In this
regard, activation of the mitogen-activated protein kinase sig-
naling pathway has been shown to inhibit adipocyte differen-
tiation (32, 33). This effect is controversial, however, because
other studies have suggested a positive role for this pathway in
adipogenesis (34, 35). In addition, the cAMP- response element-
binding protein/activating transcription factor-2 (ATF-2) and
p38 kinase signaling pathways have both been shown to be
required for PPAR
expression and efficient adipocyte differ-
entiation in 3T3-L1 cells (36 38). Interestingly, calcineurin
has been shown to affect the activity of each of these signaling
pathways in a number of distinct cell types (39 41), making
each pathway a potential target for the antiadipogenic activity
of calcineurin. Third, calcineurin may inhibit adipocyte differ-
entiation by activating a pathway that directly represses ex-
pression of PPAR
and C/EBP
. Indeed, undifferentiated
3T3-L1 preadipocytes are known to express a number of pro-
teins that have been shown to potently inhibit adipogenesis by
preventing the expression of PPAR
. The expression of these
proteins, which include PREF-1 (42), Wnt-10b (43), and the
transcription factors GATA-2 and GATA-3 (44), must be down-
regulated to ensure successful adipocyte differentiation. Inter-
estingly, calcineurin has recently been implicated in up-regu-
lating the expression of GATA-2 in muscle cell precursors,
where it is believed to play a role in the promotion of muscle cell
FIG.5.Inhibition of endogenous calcineurin activity enhances
the adipocyte differentiation of 3T3-L1 preadipocytes in re-
sponse to suboptimal adipogenic stimuli. A, 2-day, postconfluent,
3T3-L1 preadipocytes were incubated in a growth medium that con-
tained 2-fold serial dilutions of 1
MDex and 0.5 mMMix (1MD) for
2 days in the presence or absence of FK506 and thereafter in a growth
medium that contained either FK506 or vehicle control. After 10 days,
cells were stained with Oil Red O. B, 3T3-L1 preadipocytes infected
with either MSCV-VIVIT-GFP or control MSCV-H2K virus were in-
duced to undergo adipocyte differentiation with MDI in the presence or
absence of ionomycin as described under Experimental Procedures.C,
3T3-L1 preadipocytes infected with either MSCV-VIVIT-GFP or control
MSCV-H2K virus were grown for 2 days postconfluence and then in-
duced to undergo adipocyte differentiation by incubation in growth
medium that contained 2-fold serial dilutions of 1
MDex and 0.5 mM
Mix (1MD) for 2 days and maintained thereafter in growth medium
alone. After 10 days, cells were stained with Oil Red O.
Calcineurin Negatively Regulates Adipocyte Differentiation49780
by guest on November 6, 2015http://www.jbc.org/Downloaded from
differentiation and hypertrophy (45). Finally, because cal-
cineurin is best known for its ability to activate the NFAT
family of transcription factors (20, 46 48), and NFAT proteins
are expressed in 3T3-L1 cells (23), it is possible that calcineurin
mediates its effects through NFAT. Because the calcineurin-
inhibitory peptide VIVIT-GFP has previously been reported to
specifically block the ability of calcineurin to activate NFAT
proteins (22), the enhancing effect of VIVIT-GFP on adipogen-
esis (Fig. 5, Band C) suggests that NFAT proteins may play a
role in the negative regulation of adipocyte differentiation. This
notion is further supported by our observation that ectopic
expression of a constitutively active NFATc1 mutant also in-
hibits the MDI-induced differentiation of 3T3-L1 cells by pre-
venting the expression of PPAR
and C/EBP
, although this
may be secondary to a transforming effect of NFATc1 in these
cells.
2
Experiments are currently under way to further delin-
eate the role of calcineurin in the regulation of adipocyte
differentiation.
Collectively, our findings identify calcineurin as a Ca
2
-de-
pendent negative regulator of adipocyte differentiation and
provide evidence that the level of endogenous calcineurin ac-
tivity in preadipocytes plays an important role in determining
the efficiency of adipogenesis. Because it is now clear that an
increase in adipogenesis can contribute to increased adipose
tissue mass and the development of obesity (1), our data sug-
gest a potential in vivo role for calcineurin in the regulation of
obesity and its associated diseases. Indeed, our observation
that inhibition of endogenous calcineurin activity leads to en-
hanced adipogenesis may explain the increased obesity, hyper-
lipidemia, and type-2 diabetes that have been reported in pa-
tients treated with the immunosuppressive drugs CsA and
FK506 (49 51). On the basis of our results, further investiga-
tion of the in vivo role of calcineurin in the regulation of obesity
is clearly warranted.
AcknowledgmentsWe thank Janardan Reddy for the PPAR
1
cDNA and Li Liu for the generation of MSCV-VIVIT-GFP.
REFERENCES
1. Spiegelman, B. M., and Flier, J. S. (1996) Cell 87, 377389
2. Fruhbeck, G., Gomez-Ambrosi, J., Muruzabal, F. J., and Burrell, M. A. (2001)
Am. J. Physiol. Endocrinol Metab 280, E827E847
3. Visscher, T. L., and Seidell, J. C. (2001) Annu. Rev. Public Health 22, 355375
4. Rosen, E. D., and Spiegelman, B. M. (2000) Annu. Rev. Cell Dev. Biol. 16,
145171
5. Cowherd, R. M., Lyle, R. E., and McGehee, R. E., Jr. (1999) Semin. Cell Dev.
Biol. 10, 310
6. Green, H., and Kehinde, O. (1975) Cell 5, 19 27
7. Wu, Z., Bucher, N. L., and Farmer, S. R. (1996) Mol. Cell. Biol. 16, 4128 4136
8. Yeh, W. C., Cao, Z., Classon, M., and McKnight, S. L. (1995) Genes Dev. 9,
168 181
9. Rosen, E. D., Hsu, C. H., Wang, X., Sakai, S., Freeman, M. W., Gonzalez, F. J.,
and Spiegelman, B. M. (2002) Genes Dev. 16, 2226
10. Tontonoz, P., Hu, E., and Spiegelman, B. M. (1994) Cell 79, 11471156
11. Freytag, S. O., Paielli, D. L., and Gilbert, J. D. (1994) Genes Dev. 8, 1654 1663
12. Morrison, R. F., and Farmer, S. R. (1999) J. Biol. Chem. 274, 17088 17097
13. Porse, B. T., Pedersen, T. A., Xu, X., Lindberg, B., Wewer, U. M., Friis-Hansen,
L., and Nerlov, C. (2001) Cell 107, 247258
14. MacDougald, O. A., and Mandrup, S. (2002) Trends Endocrinol. Metab. 13,
511
15. Ntambi, J. M., and Takova, T. (1996) Differentiation 60, 151158
16. Miller, C. W., Casimir, D. A., and Ntambi, J. M. (1996) Endocrinology 137,
56415650
17. Shi, H., Halvorsen, Y. D., Ellis, P. N., Wilkison, W. O., and Zemel, M. B. (2000)
Physiol. Genomics 3, 7582
18. Gaillard, D., Negrel, R., Lagarde, M., and Ailhaud, G. (1989) Biochem. J. 257,
389 397
19. Vassaux, G., Gaillard, D., Ailhaud, G., and Negrel, R. (1992) J. Biol. Chem.
267, 1109211097
20. Crabtree, G. R. (2001) J. Biol. Chem. 276, 23132316
21. Clipstone, N. A., and Crabtree, G. R. (1993) Ann. N. Y. Acad. Sci. 696, 20 30
22. Aramburu, J., Yaffe, M. B., Lopez-Rodriguez, C., Cantley, L. C., Hogan, P. G.,
and Rao, A. (1999) Science 285, 2129 2133
23. Ho, I. C., Kim, J. H., Rooney, J. W., Spiegelman, B. M., and Glimcher, L. H.
(1998) Proc. Natl. Acad. Sci. U. S. A. 95, 1553715541
24. Lin, F. T., and Lane, M. D. (1992) Genes Dev. 6, 533544
25. Rosen, E. D., Sarraf, P., Troy, A. E., Bradwin, G., Moore, K., Milstone, D. S.,
Spiegelman, B. M., and Mortensen, R. M. (1999) Mol. Cell 4, 611617
26. Descombes, P., and Schibler, U. (1991) Cell 67, 569 579
27. Rusnak, F., and Mertz, P. (2000) Physiol. Rev. 80, 14831521
28. Yeh, W. C., Bierer, B. E., and McKnight, S. L. (1995) Proc. Natl. Acad. Sci.
U. S. A. 92, 11086 11090
29. Bierer, B. E., Somers, P. K., Wandless, T. J., Burakoff, S. J., and Schreiber,
S. L. (1990) Science 250, 556 559
30. Tang, Q. Q., and Lane, M. D. (1999) Genes Dev. 13, 22312241
31. Ron, D., and Habener, J. F. (1992) Genes Dev. 6, 439 453
32. Hu, E., Kim, J. B., Sarraf, P., and Spiegelman, B. M. (1996) Science 274,
2100 2103
33. Font de Mora, J., Porras, A., Ahn, N., and Santos, E. (1997) Mol. Cell. Biol. 17,
6068 6075
34. Sale, E. M., Atkinson, P. G., and Sale, G. J. (1995) EMBO J. 14, 674 684
35. Bost, F., Caron, L., Marchetti, I., Dani, C., Le Marchand-Brustel, Y., and
Binetruy, B. (2002) Biochem. J. 361, 621627
36. Engelman, J. A., Berg, A. H., Lewis, R. Y., Lin, A., Lisanti, M. P., and Scherer,
P. E. (1999) J. Biol. Chem. 274, 35630 35638
37. Reusch, J. E., Colton, L. A., and Klemm, D. J. (2000) Mol. Cell. Biol. 20,
1008 1020
38. Lee, M. Y., Kong, H. J., and Cheong, J. (2001) Biochem. Biophys. Res. Com-
mun. 281, 12411247
39. Zou, Y., Yao, A., Zhu, W., Kudoh, S., Hiroi, Y., Shimoyama, M., Uozumi, H.,
Kohmoto, O., Takahashi, T., Shibasaki, F., Nagai, R., Yazaki, Y., and
Komuro, I. (2001) Circulation 104, 102108
40. Bito, H., Deisseroth, K., and Tsien, R. W. (1996) Cell 87, 12031214
41. Lim, H. W., New, L., Han, J., and Molkentin, J. D. (2001) J. Biol. Chem. 276,
1591315919
42. Smas, C. M., Chen, L., Zhao, L., Latasa, M. J., and Sul, H. S. (1999) J. Biol.
Chem. 274, 1263212641
43. Ross, S. E., Hemati, N., Longo, K. A., Bennett, C. N., Lucas, P. C., Erickson,
R. L., and MacDougald, O. A. (2000) Science 289, 950 953
44. Tong, Q., Dalgin, G., Xu, H., Ting, C. N., Leiden, J. M., and Hotamisligil, G. S.
(2000) Science 290, 134 138
45. Musaro, A., McCullagh, K. J., Naya, F. J., Olson, E. N., and Rosenthal, N.
(1999) Nature 400, 581585
46. Clipstone, N. A., and Crabtree, G. R. (1992) Nature 357, 695697
47. Rao, A., Luo, C., and Hogan, P. G. (1997) Annu. Rev. Immunol. 15, 707747
48. Crabtree, G. R., and Olson, E. N. (2002) Cell 109 (suppl.),S67S79
49. Mathieu, R. L., Casez, J. P., Jaeger, P., Montandon, A., Peheim, E., and
Horber, F. F. (1994) Eur. J. Clin. Invest. 24, 195200
50. Abouljoud, M. S., Levy, M. F., and Klintmalm, G. B. (1995) Transplant. Proc.
27, 11211123
51. Mor, E., Facklam, D., Hasse, J., Sheiner, P., Emre, S., Schwartz, M., and
Miller, C. (1995) Transplant. Proc. 27, 1126
2
J. W. Neal and N. A. Clipstone, manuscript in preparation.
Calcineurin Negatively Regulates Adipocyte Differentiation 49781
by guest on November 6, 2015http://www.jbc.org/Downloaded from
Joel W. Neal and Neil A. Clipstone
Differentiation in 3T3-L1 Cells AdipocyteCalcium-dependent Inhibition of
Calcineurin Mediates the
TRANSDUCTION:
MECHANISMS OF SIGNAL
doi: 10.1074/jbc.M207913200 originally published online September 25, 2002
2002, 277:49776-49781.J. Biol. Chem.
10.1074/jbc.M207913200Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/277/51/49776.full.html#ref-list-1
This article cites 51 references, 27 of which can be accessed free at
by guest on November 6, 2015http://www.jbc.org/Downloaded from
... Next, we wondered what intracellular mechanisms mediated these changes. Considering the significant enriched calcium-mediated pathways in myeloid cells after GSDMD knockout, and the important role of calcium in cell differentiation [35][36][37][38], we thus assumed GSDMD deficiency-induced differentiation was calcium-related. Measuring calcium influx by flow cytometry, we observed that GSDMD deficiency attenuated calcium influx in ATRA-treated NB4 (Fig. 5G), a phenomenon that was also verified in primary GMPs isolated from NTS spleens (Fig. 5H). ...
... This leads to an uncontrolled expansion in pathogenic neutrophils, thereby exacerbating renal impairment in lupus nephritis of cell types. For example, intracellular calcium is essential for Treg cells differentiation into effector population and keratinocyte differentiation [35,36], but limits the differentiation of B cells and adipocytes [37,38]. As for myeloid progenitor cells, calcium and calcineurin signaling have been shown to inhibit cell cycle progression of GMPs to regulate myeloid cell differentiation [62]. ...
Article
Full-text available
Gasdermin D (GSDMD) is emerging as an important player in autoimmune diseases, but its exact role in lupus nephritis (LN) remains controversial. Here, we identified markedly elevated GSDMD in human and mouse LN kidneys, predominantly in CD11b ⁺ myeloid cells. Global or myeloid-conditional deletion of GSDMD was shown to exacerbate systemic autoimmunity and renal injury in lupus mice with both chronic graft-versus-host (cGVH) disease and nephrotoxic serum (NTS) nephritis. Interestingly, RNA sequencing and flow cytometry revealed that myeloid GSDMD deficiency enhanced granulopoiesis at the hematopoietic sites in LN mice, exhibiting remarkable enrichment of neutrophil-related genes, significant increases in total and immature neutrophils as well as granulocyte/macrophage progenitors (GMPs). GSDMD-deficient GMPs and all-trans-retinoic acid (ATRA)-stimulated human promyelocytes NB4 were further demonstrated to possess enhanced clonogenic and differentiation abilities compared with controls. Mechanistically, GSDMD knockdown promoted self-renewal and granulocyte differentiation by restricting calcium influx, contributing to granulopoiesis. Functionally, GSDMD deficiency led to increased pathogenic neutrophil extracellular traps (NETs) in lupus peripheral blood and bone marrow-derived neutrophils. Taken together, our data establish that GSDMD deletion accelerates LN development by promoting granulopoiesis in a calcium influx-regulated manner, unraveling its unrecognized critical role in LN pathogenesis.
... Similarly, prolonged exposure to IL-6 reduces adipogenesis and glucose transport in 3T3-F442A and 3T3-L1 cells [39]. Moreover, IL-15, despite its proinflammatory nature, mediates the calcium-dependent inhibition of adipocyte differentiation in 3T3-L1 cells by upregulating α-calcineurin [40][41][42]. ...
Article
Full-text available
Obesity stands as a significant risk factor for type 2 diabetes, hyperlipidemia, and cardiovascular diseases, intertwining increased inflammation and decreased adipogenesis with metabolic disorders. Studies have highlighted the correlation between Caspase-1 and inflammation in obesity, elucidating its essential role in the biological functions of adipose tissue. However, the impact of Caspase-1 on adipogenesis and the underlying mechanisms remain largely elusive. In our study, we observed a positive correlation between Caspase-1 expression and obesity and its association with adipogenesis. In vivo experiments revealed that, under normal diet conditions, Caspase-1 deficiency improved glucose homeostasis, stimulated subcutaneous adipose tissue expansion, and enhanced adipogenesis. Furthermore, our findings indicate that Caspase-1 deficiency promotes the expression of autophagy-related proteins and inhibits autophagy with 3-MA or CQ blocked Caspase-1 deficiency-induced adipogenesis in vitro. Notably, Caspase-1 deficiency promotes adipogenesis via Atg7-mediated autophagy activation. In addition, Caspase-1 deficiency resisted against high-fat diet-induced obesity and glucose intolerance. Our study proposes the downregulation of Caspase-1 as a promising strategy for mitigating obesity and its associated metabolic disorders.
... Cells were lysed in SDS-PAGE (sodium dodecyl sulfatepolyacrylamide gel electrophoresis) protein sample buffer and boiled at 96 • C for 10 minutes, followed by analysis on highresolution SDS-PAGE and immunoblot as described. 26 ...
Article
Full-text available
The inhibitory cell surface receptor programmed death 1 (PD1) is a major target for antibody-based cancer immunotherapies. Nevertheless, a substantial number of patients fail to respond to the treatment or experience adverse effects. An improved understanding of intracellular pathways targeted by PD1 is thus needed to develop better predictive and prognostic biomarkers. Here, via unbiased phosphoproteome analysis of primary human T cells, we demonstrate that PD1 triggering inhibited the phosphorylation and physical association with PKC theta (PKCof a variety of cytoskeleton-related proteins. PD1 blocked activation and recruitment of PKCθ to the forming immune synapse (IS) in a SHP1/SHP2 tyrosine phosphatase-dependent manner. Consequently, PD1 engagement led to impaired synaptic phosphorylation of cytoskeleton-related proteins and formation of smaller IS. TCR-induced phosphorylation of the PKCθ substrate and binding partner Vimentin was long-lasting and it could be durably inhibited by PD1 triggering. Vimentin phosphorylation in intratumoral T cells also inversely correlated with PDL1 levels in human lung carcinoma. Thus, PKCθ and its substrate Vimentin represent important targets of PD1-mediated T-cell inhibition, and low levels of Vimentin phosphorylation may serve as a biomarker for the activation of the PD1 pathway.
... Previous studies have shown that lipogenesis and adipocyte marker expression in human white preadipocytes are influenced by intracellular calcium level. 48 These effects are most likely mediated through calcium-dependent activation of calcineurin 49 and calcium/calmodulin-dependent kinase calcium levels. 50 In γ β the present study, we showed that intracellular Ca 2+ in C3H10T1/2 cells was increased by cADPR treatment and this effect was abolished by dantrolene treatment. ...
Article
Full-text available
Adipocytes play a key role in energy storage and homeostasis. Although the role of transcription factors in adipocyte differentiation is known, the effect of endogenous metabolites of low molecular weight remains unclear. Here, we analyzed time‐dependent changes in the levels of these metabolites throughout adipocyte differentiation, using metabolome analysis, and demonstrated that there is a positive correlation between cyclic adenosine diphosphate ribose (cADPR) and Pparγ mRNA expression used as a marker of differentiation. We also found that the treatment of C3H10T1/2 adipocytes with cADPR increased the mRNA expression of those marker genes and the accumulation of triglycerides. Furthermore, inhibition of ryanodine receptors (RyR), which are activated by cADPR, caused a significant reduction in mRNA expression levels of the marker genes and triglyceride accumulation in adipocytes. Our findings show that cADPR accelerates adipocytic differentiation via RyR pathway.
... The lentiCRISPRv2 vector (GeCKO) was used for CRISPR/Cas9-mediated Tensin-3 silencing. Reconstitution with Tensin-3 expression constructs was performed using a previously described retroviral vector (63). Silent point mutations and nc point mutants were generated by quick-change PCR using Kapa high-fidelity DNA polymerase (Roche), and all mutations were verified by sequencing. ...
Article
Full-text available
The protease MALT1 promotes lymphocyte activation and lymphomagenesis by cleaving a limited set of cellular substrates, most of which control gene expression. Here, we identified the integrin-binding scaffold protein Tensin-3 as a MALT1 substrate in activated human B cells. Activated B cells lacking Tensin-3 showed decreased integrin-dependent adhesion but exhibited comparable NF-κB1 and Jun N-terminal kinase transcriptional responses. Cells expressing a noncleavable form of Tensin-3, on the other hand, showed increased adhesion. To test the role of Tensin-3 cleavage in vivo, mice expressing a noncleavable version of Tensin-3 were generated, which showed a partial reduction in the T cell–dependent B cell response. Interestingly, human diffuse large B cell lymphomas and mantle cell lymphomas with constitutive MALT1 activity showed strong constitutive Tensin-3 cleavage and a decrease in uncleaved Tensin-3 levels. Moreover, silencing of Tensin-3 expression in MALT1-driven lymphoma promoted dissemination of xenografted lymphoma cells to the bone marrow and spleen. Thus, MALT1-dependent Tensin-3 cleavage reveals a unique aspect of the function of MALT1, which negatively regulates integrin-dependent B cell adhesion and facilitates metastatic spread of B cell lymphomas.
... It has been reported that calcineurin, a calcium-dependent serine/threonine phosphatase, mediates Ca 2 + dependent inhibition of adipogenesis in the early stages of differentiation in the 3T3-L1 cell line. In contrast, calcineurin has also mediated the terminal phase of adipocyte differentiation (35). Accordingly, the biphasic regulatory effect of calcium on human adipocyte differentiation has also been shown in another study (19). ...
Article
Full-text available
Background: Adipogenesis is affected by multiple factors, among which all-trans retinoic acid (ATRA) and Ca2+are considered important factors. Objectives: This study aimed to evaluate the effect of calcium, ATRA, and their underlying molecular mechanisms, alone and in combination, on adipocyte differentiation. Methods: Human adipose-derived mesenchymal stem cells (hAD-MSCs) were differentiated into adipocytes and simultaneously exposed to 0.5 μM ATRA or 2.5 mM calcium, or both in combination for 14 days. Results: Higher intracellular Ca2+was observed in both Ca2+and Ca2+plus ATRA groups. Assessment of triglyceride content and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) activity indicated lower differentiation levels in all treatment groups than in the control group. Furthermore, the results demonstrated a higher expression of retinoic acid receptor alpha (RAR-α) and lower expression of peroxisome proliferator-activated receptor γ2 (PPARγ2) and glucose transferase-4 (GLUT4) in the treatment groups as compared with the control group. It is noteworthy that Ca2+plus ATRA treatment caused more significant effects on gene expression levels (P
... It should be noted that cyanidin increased intracellular Ca 2+ in a concentration-dependent manner (10-100 μM) by activating the PLC-IP 3 pathway within seconds upon cell stimulation. This correlated with the downregulation of key adipogenic genes (PPARγ, C/EBPα, adiponectin, aP2), and agrees with the inhibitory effect of calcineurin, a Ca 2+ /calmodulin-dependent phosphatase on PPARγ and C/EBPα gene expression and adipogenesis [30]. ...
Article
Full-text available
Cyanidin is the most abundant anthocyanin found in red-purple plants and possesses anti-obesity properties. However, its mechanism of action in adipocytes remains unknown. The objective of this study was to elucidate how cyanidin inhibits adipocyte formation in 3T3-L1 preadipocytes. Cells were cultured in adipogenic differentiation medium supplemented with cyanidin and examined for adipogenesis, cell viability, and adipocyte gene expression using Oil Red O staining, MTT assay, and RT-qPCR. Real-time Ca 2+ imaging analysis was performed in living cells to elucidate cyanidin's mechanism of action. The results demonstrated that cyanidin (1-50 μM) supplementation to the adipogenic medium inhibited adipogenesis by downregulating adipogenic marker gene expression (PPARγ, C/EBPα, adiponectin, and aP2) without affecting cell viability after 4 days of treatment. Stimulation of cells with cyanidin (30-100 μM) increased intracellular Ca 2+ in a concentration dependent manner with peak calcium increases at 50 μM. Pretreatment of cells with the phospholipase C (PLC) inhibitor U73122, inositol triphosphate (IP 3) receptor blocker 2-APB, and depletion of endoplasmic reticulum Ca 2+ stores by thapsigargin abolished the Ca 2+ increases by cyanidin. These findings suggested that cyanidin inhibits adipocyte formation by activating the PLC-IP 3 pathway and intracellular Ca 2+ signaling. Our study is the first report describing the mechanism underlying the anti-obesity effect of cyanidin.
... We were first able to generate considerable amounts of highly lipid-laden murine adipocytes bearing morphological resemblance to native adipose tissues. Most studies that investigate 3T3-L1 adipogenesis in 2D are of limited culture duration (≤15 days), with lipid accumulation manifesting as small lipid droplets (Sheng et al., 2014;Neal and Clipstone, 2002;Sharma et al., 2018;Wang et al., 2013;Pazienza et al., 2016;Bogan et al., 2001;Park et al., 2018). Here, we show that extended 2D culture (30 days) of in vitro adipocytes resulted in the emergence of larger lipid droplets, a key trait of white adipose tissue. ...
Article
Full-text available
We present a method of producing bulk cell-cultured fat tissue for food applications. Mass transport limitations (nutrients, oxygen, waste diffusion) of macroscale 3D tissue culture are circumvented by initially culturing murine or porcine adipocytes in 2D, after which bulk fat tissue is produced by mechanically harvesting and aggregating the lipid-filled adipocytes into 3D constructs using alginate or transglutaminase binders. The 3D fat tissues were visually similar to fat tissue harvested from animals, with matching textures based on uniaxial compression tests. The mechanical properties of cultured fat tissues were based on binder choice and concentration, and changes in the fatty acid compositions of cellular triacylglyceride and phospholipids were observed after lipid supplementation (soybean oil) during in vitro culture. This approach of aggregating individual adipocytes into a bulk 3D tissue provides a scalable and versatile strategy to produce cultured fat tissue for food-related applications, thereby addressing a key obstacle in cultivated meat production.
Article
Obesity is a complex disorder, and the incidence of obesity continues to rise at an alarming rate worldwide. In particular, the growing incidence of overweight and obesity in children is a major health concern. However, the underlying mechanisms of obesity remain unclear and the efficacy of several approaches for weight loss is limited. As an important calcium‐permeable temperature‐sensitive cation channel, transient receptor potential vanilloid (TRPV) ion channels directly participate in thermo‐, mechano‐, and chemosensory responses. Modulation of TRPV ion channel activity can alter the physiological function of the ion channel, leading to neurodegenerative diseases, chronic pain, cancer, and skin disorders. In recent years, increasing studies have demonstrated that TRPV ion channels are abundantly expressed in metabolic organs, including the liver, adipose tissue, skeletal muscle, pancreas, and central nervous system, which has been implicated in various metabolic diseases, including obesity and diabetes mellitus. In addition, as an important process for the pathophysiology of adipocyte metabolism, adipocyte differentiation plays a critical role in obesity. In this review, we focus on the role of TRPV ion channels in adipocyte differentiation to broaden the ideas for prevention and control strategies for obesity.
Article
Enhancing meat production and quality is the eternal theme for pig breeding industries. Fat deposition has always been the focus of research in practical production because it is closely linked to pig production efficiency and pork quality. In the current study, multi-omics techniques were performed to explore the modulatory mechanisms of backfat (BF) accumulation at three core developmental stages for Ningxiang pigs. Our results identify that 15 differentially expressed genes (DEGs) and 9 significantly changed metabolites (SCMs) contribute to the BF development via the cAMP signaling pathway, regulation of lipolysis in adipocytes, and biosynthesis of unsaturated fatty acids. Herein, we found a series of candidate genes such as adrenoceptor beta 1 (ADRB1), adenylate cyclase 5 (ADCY5), ATPase Na+/K+ transporting subunit beta 1 (ATP1B1), ATPase plasma membrane Ca2+ transporting 3 (ATP2B3), ATPase Na+/K+ transporting subunit alpha 2 (ATP1A2), perilipin 1 (PLIN1), patatin like phospholipase domain containing 3 (PNPLA3), ELOVL fatty acid elongase 5 (ELOVL5) and metabolites like epinephrine, cAMP, arachidonic acid, oleic acid, linoleic acid, and docosahexaenoic acid exist age-specificeffects and play important roles in lipolysis, fat accumulation, and fatty acid composition. Our findings provide a reference for molecular mechanisms in BF tissue development and the optimization of carcass quality.
Article
Full-text available
Adipocyte differentiation is an important component of obesity and other metabolic diseases. This process is strongly inhibited by many mitogens and oncogenes. Several growth factors that inhibit fat cell differentiation caused mitogen-activated protein (MAP) kinase-mediated phosphorylation of the dominant adipogenic transcription factor peroxisome proliferator-activated receptor γ (PPARγ) and reduction of its transcriptional activity. Expression of PPARγ with a nonphosphorylatable mutation at this site (serine-112) yielded cells with increased sensitivity to ligand-induced adipogenesis and resistance to inhibition of differentiation by mitogens. These results indicate that covalent modification of PPARγ by serum and growth factors is a major regulator of the balance between cell growth and differentiation in the adipose cell lineage.
Article
Calcineurin is a eukaryotic Ca ²⁺ - and calmodulin-dependent serine/threonine protein phosphatase. It is a heterodimeric protein consisting of a catalytic subunit calcineurin A, which contains an active site dinuclear metal center, and a tightly associated, myristoylated, Ca ²⁺ -binding subunit, calcineurin B. The primary sequence of both subunits and heterodimeric quaternary structure is highly conserved from yeast to mammals. As a serine/threonine protein phosphatase, calcineurin participates in a number of cellular processes and Ca ²⁺ -dependent signal transduction pathways. Calcineurin is potently inhibited by immunosuppressant drugs, cyclosporin A and FK506, in the presence of their respective cytoplasmic immunophilin proteins, cyclophilin and FK506-binding protein. Many studies have used these immunosuppressant drugs and/or modern genetic techniques to disrupt calcineurin in model organisms such as yeast, filamentous fungi, plants, vertebrates, and mammals to explore its biological function. Recent advances regarding calcineurin structure include the determination of its three-dimensional structure. In addition, biochemical and spectroscopic studies are beginning to unravel aspects of the mechanism of phosphate ester hydrolysis including the importance of the dinuclear metal ion cofactor and metal ion redox chemistry, studies which may lead to new calcineurin inhibitors. This review provides a comprehensive examination of the biological roles of calcineurin and reviews aspects related to its structure and catalytic mechanism.
Article
The mitogenic-adipogenic activity of carbaprostacyclin (cPGI2), a stable analogue of prostacyclin (PGI2), has been proposed to be related to its ability to elicit cAMP production and to activate the protein kinase A cascade (Negrel, R., Gaillard, D., and Ailhaud, G. (1989) Biochem. J. 257, 399-405). In the present study, cPGI2 has been compared with other activators of the cAMP pathway, namely isoproterenol, forskolin and 8-bromo-cAMP, with respect to adipose cell differentiation. Carbaprostacyclin behaved as a much more potent and efficient effector of mouse Ob1771 preadipocyte differentiation than the latter agents. Moreover, cPGI2 also exerted a specific amplifying mitogenic-adipogenic role, as compared with isoproterenol in rat as well as human adipose precursor cells in primary culture, suggesting that the prostanoid was able to generate an additional second messenger. The fact that ionomycin was able to potentiate and amplify t he differentiation induced by 8-bromo-cAMP led us to give evidence, using preadipocytes preloaded with the fluorescent calcium chelator Indo-1, that cPGI2, besides its ability to activate adenyl cyclase, was also able to induce a transient increase in intracellular free calcium. This phenomenon was independent of cAMP production or inositol phospholipid breakdown and appeared to be mediated after binding to a single class of PGI2 receptor. The potential to generate simultaneously two synergistic intracellular signals allows us to ascribe to PGI2 a key and specific role in the differentiation of adipose precursor cells in vitro that would likely lead in vivo to the recruitment of "dormant" preadipocytes to become adipocytes.
Article
We have shown previously that treatment of 3T3-L1 preadipocytes with prostaglandin F2alpha (PGF2alpha) and fluprostenol, a prostanoid FP2 receptor (FP receptor) agonist, inhibited adipocyte differentiation. In this study, we demonstrate that the inhibition by PGF2alpha is controlled by concentrations of PGF2alpha rather than regulation of FP receptor levels or binding. Membranes prepared from either 3T3-L1 preadipocytes or adipocytes exhibited specific binding for PGF2alpha, suggesting that FP receptors are present throughout differentiation. Endogenous PGF2alpha production in 3T3-L1s was lower in differentiating cells compared with uninduced preadipocytes, providing further evidence that regulation occurs at the level of ligand concentration. Stimulation of the FP receptor causes a transient intracellular calcium increase, an activation of a calcium/calmodulin-dependent protein kinase (CaMK), and an increase in DNA synthesis, associated with the inhibition of differentiation. Calcium mobilizing agents, A23187 and thapsigargin, mimic the FP receptor-induced inhibition of differentiation, suggesting a role for calcium. KN-62, a CaMK inhibitor, reversed the inhibition of differentiation when added to differentiating cells with fluprostenol, suggesting a critical role for a CaMK in the inhibition. The activation of CaMK was responsible for an increase in DNA content and thymidine incorporation. The increase in DNA synthesis occurs without a concomitant increase in cell proliferation. Early differentiation markers remain intact with PGF2alpha treatment, defining the interference with normal postconfluent mitosis as the time period of differentiation that is affected by PGF2alpha. These results implicate the modulation of PGF2alpha levels in the inhibition of 3T3-L1 adipocyte differentiation through an FP receptor-mediated increase in intracellular calcium and associated increase in DNA synthesis.
Article
The flow of information from calcium-mobilizing receptors to nuclear factor of activated T cells (NFAT)–dependent genes is critically dependent on interaction between the phosphatase calcineurin and the transcription factor NFAT. A high-affinity calcineurin-binding peptide was selected from combinatorial peptide libraries based on the calcineurin docking motif of NFAT. This peptide potently inhibited NFAT activation and NFAT-dependent expression of endogenous cytokine genes in T cells, without affecting the expression of other cytokines that require calcineurin but not NFAT. Substitution of the optimized peptide sequence into the natural calcineurin docking site increased the calcineurin responsiveness of NFAT. Compounds that interfere selectively with the calcineurin-NFAT interaction without affecting calcineurin phosphatase activity may be useful as therapeutic agents that are less toxic than current drugs.
Article
The process of adipogenesis is known to involve the interplay of several transcription factors. Activation of one of these factors, the nuclear hormone receptor PPARγ, is known to promote fat cell differentiation in vitro. Whether PPARγ is required for this process in vivo has remained an open question because a viable loss-of-function model for PPARγ has been lacking. We demonstrate here that mice chimeric for wild-type and PPARγ null cells show little or no contribution of null cells to adipose tissue, whereas most other organs examined do not require PPARγ for proper development. In vitro, the differentiation of ES cells into fat is shown to be dependent on PPARγ gene dosage. These data provide direct evidence that PPARγ is essential for the formation of fat.
Article
Calcium signaling activates the phosphatase calcineurin and induces movement of NFATc proteins into the nucleus, where they cooperate with other proteins to form complexes on DNA. Nuclear import is opposed by kinases such as GSK3, thereby rendering transcription continuously responsive to receptor occupancy. Disruptions of the genes involved in NFAT signaling are implicating this pathway as a regulator of developmental cell–cell interactions.
Article
When cells of the established preadipose line 3T3-L1 enter a resting state, they accumulate triglyceride and convert to adipose cells. The adipose conversion is brought about by a large increase in the rate of triglyceride synthesis, as measured by the incorporation rate of labeled palmitate, acetate, and glucose. In a resting 3T3 subline which dose not undergo the adipose conversion, the rate of triglyceride synthesis from these precursors is very low, and similar to that of growing 3T3-L1 cells, before their adipose conversion begins. If 3T3-L1 cells incorporate bromodeoxyuridine during growth, triglyceride synthesis does not increase when the cells reach a stationary state, and triglycerides do not accumulate. As would be expected from their known actions on tissue adipose cells, lipogenic and lipolytic hormones and drugs affect the rate of synthesis and accumulation of triglyceride by 3T3-L1 cells, but in contrast to bromodeoxyuridine, these modulating agents do not seem to affect the proportion of cells which undergoes the adipose conversion. Insulin markedly increases the rate of synthesis and accumulation of triglyceride by fatty 3T3-L1 cells, and produces a related increase in cell protein content. Of 20 randomly selected clones isolated from the original 3T3 stock, 19 are able to convert to adipose cells. The probability of such a conversion varies greatly among the different clones, in most cases being much lower than for 3T3-L1; but once the conversion takes place, the adipose cells produced from all of the 19 clones appear similar. The adipose conversion would seem to depend on an on-off switch, which is on with a different probability in different clones. This probability is quasistably inherited by the clonal progeny.