Вы находитесь на странице: 1из 16

Article

Triglyceride Synthesis by DGAT1 Protects


Adipocytes from Lipid-Induced ER Stress during
Lipolysis
Graphical Abstract Authors
Chandramohan Chitraju,
Adipocyte
during Niklas Mejhert, Joel T. Haas, ...,
Lipid Droplet fasting Suneil K. Koliwad, Tobias C. Walther,
Robert V. Farese, Jr.
TG
FA Correspondence
twalther@hsph.harvard.edu (T.C.W.),
robert@hsph.harvard.edu (R.V.F.)
DGAT1
Wild type In Brief
Chitraju et al. unravel a decades-old
mystery of why a large portion of
hydrolyzed FAs in adipocytes is re-
Lipid Droplet
esterified to TGs during lipolysis. They
show that the ER enzyme DGAT1
TG FA mediates this FA re-esterification, not to
FA FA
preserve TG mass but instead to protect
the ER from lipotoxicity.
ER-Stress
DGAT1 Knockout

Highlights
d DGAT-mediated triglyceride synthesis prevents lipid-
induced ER stress

d During lipolysis, DGAT1 is upregulated and mediates fatty


acid re-esterification

d DGAT1 activity protects adipocytes from lipid-induced ER


stress during lipolysis

Chitraju et al., 2017, Cell Metabolism 26, 407–418


August 1, 2017 ª 2017 Elsevier Inc.
http://dx.doi.org/10.1016/j.cmet.2017.07.012
Cell Metabolism

Article

Triglyceride Synthesis by DGAT1


Protects Adipocytes from Lipid-Induced
ER Stress during Lipolysis
Chandramohan Chitraju,1,2 Niklas Mejhert,1,2 Joel T. Haas,5,8 L. Grisell Diaz-Ramirez,5,9 Carrie A. Grueter,5,10
Jason E. Imbriglio,6 Shirly Pinto,6 Suneil K. Koliwad,5,7,11 Tobias C. Walther,1,2,3,4,12,* and Robert V. Farese, Jr.1,2,3,12,13,*
1Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
2Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
3Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
4Howard Hughes Medical Institute, Boston, MA 02115, USA
5Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
6Merck & Co., Inc., Kenilworth, NJ 07033, USA
7Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
8Present address: Université Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, F-59000 Lille, France
9Present address: Division of Geriatrics, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
10Present address: Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
11Present address: Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
12These authors contributed equally
13Lead Contact

*Correspondence: twalther@hsph.harvard.edu (T.C.W.), robert@hsph.harvard.edu (R.V.F.)


http://dx.doi.org/10.1016/j.cmet.2017.07.012

SUMMARY tissue (WAT). However, when adipose depots of TG are over-


whelmed, for instance during obesity, other cells will synthesize
Triglyceride (TG) storage in adipose tissue provides and accumulate TG in LDs. This is often accompanied by tissue
the major reservoir for metabolic energy in mammals. dysfunction due to cellular lipotoxicity (Unger and Zhou,
During lipolysis, fatty acids (FAs) are hydrolyzed from 2001), which can lead to lipotoxicity-related diseases, such as
adipocyte TG stores and transported to other tissues type 2 diabetes, non-alcoholic fatty liver disease, and metabolic
for fuel. For unclear reasons, a large portion of hydro- cardiomyopathy.
When energy supply is limited, lipolysis of adipocyte TGs is
lyzed FAs in adipocytes is re-esterified to TGs in a
stimulated, releasing large amounts of hydrolyzed FAs that can
‘‘futile,’’ ATP-consuming, energy dissipating cycle.
be transported to other tissues for use as fuel (Duncan et al.,
Here we show that FA re-esterification during adipo- 2007; Zechner et al., 2012). However, during lipolysis, up to
cyte lipolysis is mediated by DGAT1, an ER-localized two-thirds of hydrolyzed FAs from TG is re-esterified to TG in
DGAT enzyme. Surprisingly, this re-esterification WAT (Edens et al., 1990; Leibel et al., 1985; Nye et al., 2008;
cycle does not preserve TG mass but instead func- Vaughan, 1962; Wood et al., 1960). Re-esterification of FAs to
tions to protect the ER from lipotoxic stress and glycerol backbones could occur at any step along the glyceroli-
related consequences, such as adipose tissue pid-TG synthesis pathway. This re-esterification cycle is tar-
inflammation. Our data reveal an important role for geted by thiazolidinedione drugs, which promote glyceroneo-
DGAT activity and TG synthesis generally in averting genesis (Guan et al., 2002; Tordjman et al., 2003) to provide
ER stress and lipotoxicity, with specifically DGAT1 glycerol and glycerol-phosphate for TG synthesis during fasting
and which promote fat storage in humans. The reason for this
performing this function during stimulated lipolysis
‘‘futile,’’ ATP-consuming re-esterification cycle has remained a
in adipocytes.
mystery for decades. For instance, this cycle of lipolysis and
re-esterification might provide a mechanism to fine-tune the
release rate of FAs, thereby preserving fat mass during fasting
INTRODUCTION or other conditions associated with lipolysis, but evidence for
this hypothesis is lacking.
Triglycerides (TGs) provide the major storage form of fatty acids The enzyme responsible for re-esterification of FAs to diacyl-
(FAs), which in turn serve as reservoirs of metabolic fuel or mem- glycerol to form TGs during adipocyte lipolysis is unknown. In
brane building blocks. During times of excess energy availability, mammals, TG synthesis is catalyzed by acyl CoA:diacylglycerol
more TG is synthesized, and in mammals this TG is predomi- acyltransferase (DGAT) enzymes, DGAT1 or DGAT2 (Liu et al.,
nantly packaged in cellular lipid droplets (LDs), the organelle 2012; Yen et al., 2008). Both DGAT enzymes catalyze the
for neutral lipid storage (Farese and Walther, 2009; Fujimoto same reaction, utilizing diacylglycerol and fatty acyl CoAs as
and Parton, 2011; Walther and Farese, 2012). TG storage substrates, but are evolutionarily unrelated (Liu et al., 2012;
in mammals occurs primarily in adipocytes in white adipose Yen et al., 2008). DGAT1 is localized exclusively to the ER (Stone

Cell Metabolism 26, 407–418, August 1, 2017 ª 2017 Elsevier Inc. 407
A ad lib 16h fast 4h re-fed B LDs mediates the FA re-esterification cycle in adipocytes is
unclear.
Gonadal fat ** DGAT1
** Here, using a combination of cell and murine models, we
200 investigated how TG synthesis is mediated in adipocytes during
2
** *** lipolysis. We found that the re-esterification cycle is mediated
** ***
exclusively by DGAT1 and that DGAT2 is inactive during these
1 *** 100 ***
*** conditions. Surprisingly, we found that the function of DGAT1-

Activity [A.U]
mediated re-esterification during lipolysis is not to preserve TG
0 0 mass, but instead DGAT1 activation serves to protect the adipo-
Brown fat DGAT2 cyte ER from lipotoxic stress and associated inflammatory con-
Relative mRNA

*** *** sequences. Our findings thus uncover that TG synthesis is


** *** ** 150
2 important to protect cells from lipid-induced ER stress.
***
***
RESULTS
1 75
DGAT1 and DGAT2 in Murine WAT Are Reciprocally
0 0 Regulated in Fasting and Re-feeding
Liver
d
st

We investigated mRNA regulation of DGAT1 and DGAT2 in


-fe
fa

***
re
h

3 *** gonadal WAT, brown adipose tissue (BAT), and livers from
16

4h

mice in different nutritional states. In WAT, DGAT2 mRNA levels


2 were decreased after 16 hr fasting and increased after 4 hr of re-
*** feeding (Figure 1A). These expression changes closely mirrored
1 those of sterol regulatory element binding protein 1c (SREBP1c)
0 n.d. and leptin (Figure 1A), two known master regulators of the WAT
response to changes in food intake. Consistent with changes in
in

2
P1

AT
pt

AT

mRNA levels, DGAT2 enzymatic activity in WAT was increased


Le

EB

during re-feeding (Figure 1B). In contrast, DGAT2 mRNA levels


D

D
SR

in liver and BAT remained unregulated during fasting and re-


feeding. Unexpectedly, the level of DGAT1 mRNA was increased
Figure 1. DGAT1 and DGAT2 in Murine WAT Are Reciprocally Regu-
lated in Fasting and Re-feeding
during 16 hr fasting, a physiological condition associated with
(A) Relative mRNA levels in gonadal fat, brown fat, and livers of mice that were increased lipolysis and reduced TG synthesis (Figure 1A).
fed ad libitum, fasted for 16 hr, or after 16 hr fasted and 4 hr re-fed. Cyclophilin Upon re-feeding, DGAT1 mRNA levels normalized to the levels
was used as reference gene. observed during ad libitum feeding conditions. These changes
(B) DGAT1 and DGAT2 enzymatic activities were measured in microsomal in mRNA levels were reflected in increased enzymatic DGAT1
fractions of WAT from wild-type mice and DGAT1 KO mice, respectively. Data
activity during fasting (Figure 1B). The upregulation of DGAT1
are presented as ± SD (n = 5 mice per group). Statistical significance was
evaluated by unpaired two-tailed Student’s t test. **p < 0.01; ***p < 0.001; n.d.,
was not only observed in WAT but also in liver, as reported
not detected. (Villanueva et al., 2009), and to some extent in BAT.

DGAT1 Mediates the Majority of TG Re-esterification


et al., 2009; Wilfling et al., 2013) and has broader substrate spec- during Stimulated Lipolysis In Vitro
ificity (with respect to acyl acceptors) than DGAT2 (Cases et al., Our data indicate that DGAT1-mediated TG synthesis has a
1998, 2001; Yen et al., 2005, 2008), and mice lacking DGAT1 are more active role in WAT during fasting than during fed condi-
viable and metabolically healthy (Chen et al., 2002a; Smith et al., tions, suggesting that DGAT1 mediates FA re-esterification
2000; Yen et al., 2008). In contrast, DGAT2 localizes both to the during this condition. To test this hypothesis, we studied differ-
ER and around LDs (Stone et al., 2009; Wilfling et al., 2013) and entiated 3T3-L1 adipocytes and examined TG synthesis by
appears to be the major enzyme accounting for TG synthesis DGAT1 or DGAT2 under different conditions. Under basal condi-
(Cases et al., 2001; Stone et al., 2004), as mice lacking the tions, inhibiting either DGAT1 or DGAT2 only modestly reduced
enzyme have >90% reduction in TGs and die shortly after birth TG synthesis in intact cells (measured by [14C]-oleic acid
(Stone et al., 2004). DGAT1 and DGAT2 have been functionally incorporation), but inhibiting both DGAT1 and DGAT2 nearly
linked to storage of TGs in different types of LDs. TG synthesis completely abolished TG synthesis (Figure 2A). Under these
mediated by DGAT1 in the ER leads exclusively to the formation conditions, there was a concomitant increase in label in cellular
of relatively small (400–800 nm diameter) LDs, called initial 1,2-DAG (or a racemic mixture of sn-1,2 DAG and sn-2,3 DAG
LDs (or iLDs; Wilfling et al., 2013). In contrast, when FA levels isomers) and free FAs. These findings suggest that DGAT1 and
are high in cells, DGAT2 re-localizes from the ER around a subset DGAT2 activities compensate for each other to maintain TG
of LDs (Kuerschner et al., 2008; Stone et al., 2009; Wilfling synthesis in adipocytes under basal conditions.
et al., 2013), where it acts in concert with other LD-localized We next examined TG synthesis during lipolysis (stimulated
enzymes of TG synthesis to locally synthesize TGs and with 10 mM isoproterenol). Using labeled FA as tracer, we found
generate expanding LDs (or eLDs, typically 1–2 mm in diameter). that a considerable amount of TG was synthesized at a 3 hr time
Which DGAT enzyme and related pathway of TG synthesis for point, consistent with previous reports on FA re-esterification

408 Cell Metabolism 26, 407–418, August 1, 2017


SO Figure 2. DGAT1 Mediates the Majority of

SO
2i

2i
1D

1D
TG Re-esterification during Stimulated
M

M
1i

2i

1i

2i
A B FA in medium

D
D

D
D

D
210 Lipolysis In Vitro

CpmX104/mg protein
TG (A) Incorporation of [14C] oleic acid into TG in
***
** differentiated 3T3-L1 adipocytes was measured
140 during basal and 3 hr stimulated lipolysis by in-
1,3-DAG FA hibiting DGAT1 (D1i) or DGAT2 (D2i) or both
1,2/2,3- (D1D2i). Lipids were extracted from cells and
DAG Basal Lipolysis Stimulated Lipolysis 70
separated by TLC. (Top) Autoradiographs of TLC
1800 TG plates. Lipids from TLC plates were scraped out
0 and quantified by scintillation counter. Data shown
900 TLC FA represent two independent experiments.
(B) FA release into medium from differentiated 3T3-
DMSO D1i D2i D1D2i
*** *** L1 adipocytes was measured by extracting lipids
0 *** from the medium and separating them by TLC. FA
*** FA C FA in medium
200 region on TLC (shown in the graph) was scraped
200
off, and radioactivity was measured by liquid
CpmX102/mg protein

100 *** ***


CpmX103/mg protein
scintillation counter.
***
50 (C) FA release into medium from differentiated 3T3-
L1 adipocytes was measured by counting radio-
***
0 100 activity in the medium (in B, we used a higher
300 *** quantity of tracer. In subsequent experiments, we
1,2/2,3-DAG
reduced tracer amount to label cells). Data are
*** presented as ± SD (n = 3 biological replicates).
150
Statistical significance shown is between DMSO
*** ***
0 control treatments and corresponding inhibitor
0 treatments. Statistical significance was evaluated
ATGLi + + + +
100 1,3-DAG HSLi + + + + by unpaired two-tailed Student’s t test. **p < 0.01;
***p < 0.001. See also Figures S1 and S2.
DGAT1i + +
DGAT2i + +
50
*** ***
0 into the medium. Consistent with this,
SO

SO
2i

2i

we observed a roughly 2-fold increase


1i

D 2i
1i

D 2i
1D

1D
D
D
D
D
M

M
D

of FA release from 3T3-L1 adipocytes


when DGAT1 was inhibited during lipol-
during lipolysis (Edens et al., 1990; Leibel et al., 1985; Nye et al., ysis (Figure 2B). In contrast, no changes in FA release were found
2008; Vaughan, 1962). Notably, DGAT1 inhibition nearly abol- upon inhibition of DGAT2. Inhibition of both DGAT1 and DGAT2
ished TG synthesis during stimulated lipolysis (Figure 2A), and resulted in effects similar to those for DGAT1 inhibition alone.
free FAs and 1,2-diacylglycerol accumulated in cells. DGAT1 in- The increase of released FAs from adipocytes with DGAT1 in-
hibition also resulted in accumulation of monoacylgycerol (Fig- hibition during lipolysis could result from either an increase in
ure S2B). In contrast, DGAT2 inhibition alone had no effect on lipolysis activity or a decrease in re-esterification of FAs liberated
[14C]-oleic acid incorporation into TG during stimulated lipolysis. from TG. To examine these possibilities, we stimulated lipolysis
Also, inhibiting DGAT2 in addition to DGAT1 did not change the in adipocytes under conditions in which we inhibited the major
findings for DGAT1 inhibition alone, indicating that DGAT2 is adipocyte lipases. Consistent with previous reports, inhibition
apparently not active during stimulated lipolysis. of adipose triglyceride lipase (ATGL), the rate-limiting enzyme
To exclude the possibility that the reduced TG synthesis found of adipocyte TG hydrolysis, reduced FAs release from cells by
with DGAT1 inhibition was due to increased dilution of the tracer 80% (Figure 2C). Inhibition of DGAT1, but not DGAT2, under
([14C] oleic acid) from release of hydrolyzed FAs during lipolysis, these conditions greatly increased FA release into the medium.
we examined tracer incorporation into TG and other lipids for a Blocking hormone-sensitive lipase (HSL) alone reduced FA
range of tracer concentrations. We found that the incorporation release to a lesser degree than ATGL inhibition. Similar to
of the tracer into PC and PE in DMSO-treated or DGAT1-in- ATGL inhibition, however, inhibition of DGAT1, but not DGAT2,
hibited cells was linearly increased and similar for each tracer resulted in increased FA release (Figure 2C). These data suggest
specific activity (Figures S1A and S1B), arguing against a signif- that increased FA release during DGAT1 inhibition is due to a
icant tracer dilution effect accounting for the findings in DGAT1- block in DGAT1 mediated re-esterification rather than changes
inhibited cells. The incorporation of tracer into TG also increased to in lipolysis activity per se. Consistent with this interpretation,
under the DGAT1-inhibition conditions, although at a slightly we did not observe any changes in ATGL and HSL protein levels
lower proportion than in the control cells, perhaps because TG or HSL phosphorylation with DGAT1 inhibition (Figure S2A).
synthesis was nearly absent.
Together, these data indicate that DGAT1 mediates most TG DGAT1 Forms Initial LDs in Cultured Adipocytes during
synthesis under stimulated lipolysis conditions in cultured adipo- Stimulated Lipolysis
cytes. This conclusion predicts that DGAT1 inhibition during Under conditions of stimulated lipolysis, if FAs are not seques-
isoproterenol-stimulated lipolysis leads to increased FA release tered from cells by incubating with albumin in the medium,

Cell Metabolism 26, 407–418, August 1, 2017 409


A Basal ISO ISO+Triacsin C Figure 3. DGAT1 Forms iLDs during Stimu-
lated Lipolysis
(A) Confocal microscopic images of differentiated
3T3-L1 cells in basal and isoproterenol stimulated
lipolysis (ISO). Lipolysis was induced by treating
cells with 10 mM isoproterenol in the medium con-
taining no serum and no BSA. LDs were stained with
BODIPY 493/503. Scale bar, 10 mm.
(B) LDs were quantified using IMARIS software.
Representative results from four independent ex-
periments are shown.

determine whether this was also the case


in in vivo, we performed studies of mice
lacking DGAT1 specifically in adipocytes.
To avoid complications due to altered
whole-body physiology and cross-talk be-
tween tissues observed in mice lacking
DGAT1 in all tissues (Chen et al., 2002a;
ISO+D1i ISO+D2i ISO+D1D2i Smith et al., 2000; Yen et al., 2008), we
generated mice lacking DGAT1 specifically
B in adipocytes (ADGAT1 KO) by crossing
10-20 μm Dgat1Flox/Flox mice (Villanueva et al., 2009)
LD diameter [μm]

2-10 μm 15 with transgenic mice expressing Cre-re-


2000
LD number

<1 μm
combinase under the adiponectin pro-
1500 5 moter (Adipoq-Cre; Eguchi et al., 2011).
100 ADGAT1 KO mice lacked detectable
3 DGAT1 protein and had markedly reduced
50
1 DGAT activity in adipose tissue (under con-
0 ditions optimized to detect DGAT1 activity)
but had normal DGAT1 expression in the
ia O

SO
C

C
1i
2i

1i
2i
D al

l
sa
S

D
D

D
D
s

in

in

liver and small intestine (Figures 4A, 4B,


Ba

Ba
M

M
cs

cs
D
ia

and S3A).
Tr

Tr

ADGAT1 KO mice fed a chow diet


+ ISO + ISO
ad libitum had smaller adipose tissue
depots (20% less weight than controls)
3T3-L1 cells form larger numbers of new iLDs (Ariotti et al., 2012; (Figure S3B) and had similar levels of plasma glucose, TG,
Hashimoto et al., 2012; Marcinkiewicz et al., 2006; Paar et al., glycerol, and FAs under standard ad libitum-fed conditions
2012). Inasmuch as we found that DGAT1 is specifically active (Figure 4C). The lack of increase in plasma FAs in the fasted
during lipolysis, we tested whether the new iLDs are formed by ADGAT1 KO mice was unexpected and could be due to
the DGAT1-coupled iLD pathway. Consistent with previous find- increased oxidation of FAs in tissues, since these mice exhibit
ings (Ariotti et al., 2012; Paar et al., 2012), we detected by increased energy expenditure (C.C., T.C.W., and R.V.F., unpub-
confocal microscopy a multitude of relatively uniformly sized, lished data). ADGAT1 KO mice also showed no differences in
small (median diameter 0.75 mm) iLDs that were newly formed liver TG levels after fasting (Figure S3C). To test the role of
when lipolysis was triggered in 3T3-L1 adipocytes (Figure 3A). DGAT1 during lipolysis in adipocytes of adipose tissue, we
Treatment with Triacsin C, which blocks the activation of FAs measured glycerol release from isoproterenol-treated WAT ex-
with a coenzyme A moiety and hence the incorporation of FAs plants of control or ADGAT1 KO fat pads. At baseline and during
into glycerolipids, effectively blocked the formation of these lipolysis, we found that ADGAT1 KO fat released more glycerol
newly formed iLDs, as reported (Paar et al., 2012). Testing the into the medium than fat pads isolated from control mice
roles of the two different DGAT enzymes in this process, we (Figure 4D). Increased release of glycerol and FAs was also
found that inhibition of DGAT1, but not DGAT2, blocked the for- observed in WAT explants treated with DGAT1 inhibitor (Fig-
mation of newly formed iLDs (see Figure 3B, in particular LDs ure 4E), consistent with a role for DGAT1 in re-esterification dur-
with <1 mm diameter). ing lipolysis.

Decreased Re-esterification in Cultured WAT from Mice DGAT1-Mediated Re-esterification Does Not Preserve
Lacking DGAT1 in Adipocytes Fat Mass during Fasting or Calorie Restriction
Our findings in cultured 3T3-L1 adipocytes show that DGAT1, Identification of DGAT1 as the enzyme responsible for cata-
but not DGAT2, re-esterifies FAs during stimulated lipolysis. To lyzing FA re-esterification during lipolysis in adipocytes, both

410 Cell Metabolism 26, 407–418, August 1, 2017


A C Controls ADGAT1 KO Figure 4. DGAT1-Mediated Re-esterification
WAT Liver Does Not Prevent Fat Loss during Fasting or

Glucose (mg/dl)
DGAT1 120 80 Calorie Restriction in Mice

TG (mg/dl)
90 (A) Western blot analysis showing absence of DGAT1
Tubulin 60
protein in WAT lysates of ADGAT1 KO mice.
60 40
(B) DGAT1 activity in WAT microsomal fractions of
Oo ols
ls O
1 K ontr
ntr 1K 30 20 control and ADGAT1 KO mice.
Co G AT C G AT 0 0
AD AD 16h fast 4h re-fed 16h fast 4h re-fed
(C) Plasma glucose, glycerol, free FA, and TG of 16 hr
fasted and 4 hr re-fed mice (n = 5–6 mice per group).
B DGAT1 activity - WAT (D) Glycerol release from WAT explants under basal

Glycerol (m mol/l)
NEFA (m mol/l) and 3 hr stimulated (ISO) lipolysis conditions.
TG 2.0 0.4
(E) Glycerol and FA release from WAT explants treated
Activity [A. U.]

1.5 0.3 with DGAT inhibitors under stimulated (ISO) lipolysis for
200
1.0 0.2 3 hr.
100 0.5 0.1 (F) Body weights and fat mass loss during 16 hr fasting
*** 0.0 0.0 (n = 5–6 mice per group).
0
16h fast 4h re-fed 16h fast 4h re-fed (G and H) Body weights and change in lean and
Controls ADGAT1 KO
fat mass during 60% calorie restriction study for
D E
Controls ADGAT1 KO Glycerol FFA 20 days, followed by recovery by ad libitum feeding (n =
8–10 mice per group). Data are presented as ±SD.
(n mol/mg protein)

*
(n mol/mg protein)

400 Glycerol DMSO * * *


* 600 D1i Statistical significance was evaluated by unpaired two-
D2i tailed Student’s t test. *p < 0.05; ***p < 0.001. See also
200 400 D1D2i Figure S3.
*
200
0 0
Basal ISO Basal ISO Basal ISO

F Body weight Lean mass DGAT1 Protects against the ER


% of gonadal fat loss
during 16 h fasting

Stress Response during Simulated


20
20 Controls
Mass (g)

40 Lipolysis in 3T3-L1 Adipocytes


ADGAT1 KO ER function is closely monitored by a ho-
10
10 20 meostatic system, termed the unfolded
0 protein response (UPR), which is activated
0
ad lib 16h fast ad lib 16h fast during the accumulation of misfolded pro-
teins, during an overload of the ER with
G H secretory cargo, or during changes in
35 Controls Lean mass Fat mass
ADGAT1 KO 30 the lipid composition of the ER (Volmer
Body weight (g)

30
4 and Ron, 2015; Walter and Ron, 2011).
20
Mass (g)

25 3 DGAT1 has been functionally linked to pre-


2 venting cellular lipotoxicity (Koliwad et al.,
20 10
1 2010; Listenberger et al., 2003). We there-
15 0 0 fore hypothesized that DGAT1 activation
0 2 4 10 20 1 2 7 13 Day 0 Day 20 Day 0 Day 20 during lipolysis protects the ER from lipo-
60% Calorie restriction Recovery toxic stress due to the large increase in
FAs. The UPR in mammals consists of
three branches, employing distinct stress
in culture and in WAT, enabled us to next address the sensors (IRE1, PERK, and ATF6) and response programs (Walter
function of this process. One possibility is that DGAT1-medi- and Ron, 2011).
ated re-esterification during lipolysis prevents loss of fat We first tested for UPR activation in differentiated 3T3-L1
mass when energy input is limited. To test this hypothesis, adipocytes during basal conditions. Although inhibiting
we submitted ADGAT1 knockout mice to nutrient deprivation DGAT1 or DGAT2 alone did not trigger the UPR, inhibiting
caused either by fasting or by calorie restriction. During them simultaneously resulted in strong induction (e.g., 80-
conditions of a 16 hr fast, both ADGAT1 and control mice fold induction of Xbp1s after 10 hr of inhibition) of many UPR
lost similar amounts of body weight, lean mass, and fat targets genes, including spliced Xbp1, Bip, Chop, and Atf3
mass (Figure 4F). Similarly, during calorie restriction for (Figures 5A, 5B, and 5E). These changes were similar in magni-
20 days with a diet consisting of 40% of normal food intake tude to those found with treatment with thapsigargin, an agent
(60% calorie restriction; Zhao et al., 2010), body weights and that induces ER stress by blocking calcium uptake and
reductions in lean and fat masses were similar between geno- thereby disrupting ER homeostasis. If the UPR is unresolved,
types (Figures 4G and 4H). These data suggest that DGAT1- this leads to cleavage and degradation of messages encoding
mediated re-esterification of FAs does not play a major role some ER proteins, a process known as RIDD, or regulated
in preserving fat mass during conditions of acute or chronic Ire1-dependent decay of mRNAs (Han et al., 2009; Hollien
energy deprivation. et al., 2009; Hollien and Weissman, 2006; Maurel et al.,

Cell Metabolism 26, 407–418, August 1, 2017 411


A IRE1α/PERK branch targets RIDD
B
80 Xbp1s ***
*** 12 Chop ***

39a
40

1
1
8

90b

24d
pud

jb1

nat
***

frb
1s
1t

em
a5
p
Atf3
Atf4

Dna
Xbp
Xbp

Hsp
2

Pdg
Hgs
Cho

Sec

Relative mRNA
Her
B ip

Pdi
Tm
4
D1i 0 0
D2i 18 Bip *** 120 Atf3 ***
D1D2i 12 *** 60
***
Tg
2 4
-5 -2 0 3 5
0 0
log 2

Tg
SO
Tg

D 2i
2i
D 2i

1i
2i
SO

1i

D
1D
D

D
1D
D

M
M

D
D
C IRE1α/PERK branch targets RIDD D
25 Xbp1s *** DMSO
4 Chop
39a

D1i
1
1

***
90b

24d
pud

jb1

nat

16 *** *** ***


frb
1s
1t

em

D2i
a5
p

***
Atf3
Atf4

Dna
Xbp
Xbp

Hsp

Pdg
Hgs
Cho

Sec

***
Her
Bip

Pdi
Tm

8 2 D1D2i

Relative mRNA
30 min Tg
D1i ** 0
3h 0
10 Bip *** 4 Atf3 ***
***
30 min ***
D2i 4
3h ***
*** 2
2
30 min
D1D2i
3h
0 0
30 min 30 min 3h 30 min 3h
Tg
3h
E Basal Lipolysis Stimulated Lipolysis
-5 -2 0 3 5 Bip

log 2 CHOP
Tubulin

SO 1i 2i 2i Tg SO 1i 2i 2i Tg
M D D 1D M D D 1D
D D D D

Figure 5. DGAT1 Inhibition during Simulated Lipolysis Induces the ER Stress Response in 3T3-L1 Adipocytes
(A and B) Heatmap and bar graphs showing relative mRNA levels of ER stress marker genes during basal lipolysis determined by RT-qPCR after 10 hr treatment
with DGAT1 (D1i), DGAT2 (D2i), or both (D1D2i) inhibitors. Thapsigargin (Tg)-treated cells were used as positive controls.
(C and D) Heatmap and bar graphs showing relative mRNA levels of ER stress marker genes during induced lipolysis. Lipolysis was induced by treating cells with
10 mM isoproterenol in the medium containing no serum and no BSA.
(E) Western blot analysis of Bip and CHOP during basal and induced lipolysis. Data are presented as ±SD (n = 3 biological replicates). Statistical significance
shown is between DMSO control treatments and corresponding inhibitor treatments. Statistical significance was evaluated by unpaired two-tailed Student’s
t test. ***p < 0.001. See also Figures S4 and S5.

2014). Indeed, we found decreased abundance of RIDD target FAs from either de novo synthesis or TG hydrolysis can lead to
mRNAs during prolonged inhibition of both DGAT1 and DGAT2 an ER stress response.
under basal conditions, findings that were also consistent with During lipolysis in adipocytes, our studies show that TG syn-
those after thapsigargin treatment. These data indicate that thesis activity by DGAT2 appears to be inactivated (Figure 2A).
under basal conditions in differentiated adipocytes, either We thus hypothesized that activation of DGAT1 functions during
DGAT1 or DGAT2 activity is sufficient to protect the ER from lipolysis primarily to re-esterify FAs in the ER and prevent disrup-
lipotoxic stress, but in the absence of TG synthesis, the UPR tion of ER function. Indeed, during stimulated lipolysis, inhibition
and ER stress are markedly activated. The increase in ER of DGAT1 alone (or DGAT1 inhibition combined with DGAT2 in-
stress caused by inhibition of both DGAT1 and DGAT2 in the hibition) triggered strong induction of the UPR (16-fold induction
basal conditions was partially blocked by pharmacologic inhi- of Xbp1s after 3 hr of DGAT1 inhibition) and reduced abundance
bition of FA synthesis (data not shown), suggesting that excess of RIDD targets (Figures 5C–5E). These effects were dependent

412 Cell Metabolism 26, 407–418, August 1, 2017


on the generation of non-esterified FAs, as increasing the extra- DGAT1- or DGAT2-inhibition conditions. As we found in murine
cellular concentration of albumin, which binds to and effectively cells, the mRNA levels of ER stress genes XBP1s and Bip were
buffers excess FAs, decreased the magnitude of UPR activation increased in cells treated with both DGAT inhibitors under basal
(Figure S4). However, we found no effect on ER function as conditions, whereas these markers were elevated with DGAT1
measured by the amount of proteins secreted by differentiated inhibition alone with stimulated lipolysis (Figure S7), validating
adipocytes into the medium (Figure S5). As expected from the this response for human adipocytes.
observation that DGAT2 was inactive during lipolysis, inhibition To further examine the relationship between DGAT1 expres-
of DGAT2 did not result in induction of the UPR or RIDD under sion and the expression of ER stress genes in human adipose tis-
these conditions (Figures 5C–5E). sue in pathophysiological conditions, we analyzed published
mRNA expression profiles of human WAT obtained from 26
Lack of DGAT1 in Adipocytes Increases the Unfolded non-obese and 30 obese women who were fasted overnight (Ar-
Protein Response in WAT during Fasting or Cold ner et al., 2012). Expression levels of DGAT1 and DGAT2 and
Exposure genes regulated by the UPR (GO: 0006986) were extracted
To test whether DGAT1 protects the ER from lipotoxic stress and from the dataset and correlated. We found that the abundance
UPR activation in vivo, we studied conventionally housed wild- of numerous mRNAs encoded by UPR genes displayed a nega-
type or ADGAT1 KO mice under two conditions that stimulate tive correlation with DGAT1 mRNA levels (Figure 7A). This
lipolysis in WAT: a 16 hr fast or 6 hr exposure to cold in the type of relationship with UPR genes was not found for DGAT2.
absence of food. Even at basal conditions, we found a small, The mRNA levels for six genes that correlate negatively with
but significant, activation of Xbp1s and the ER chaperone Bip DGAT1 are shown for different individuals in Figure 7B. Taken
in WAT of ADGAT1 compared with controls (Figure 6A). After together, these data corroborate the apparent relationship of
16 hr of fasting, or after cold exposure, several UPR targets DGAT1 to ER stress in human adipose tissue.
(e.g., Xbp1s and Bip) were upregulated 2- to 3-fold in WAT.
In contrast, mRNA levels of Atf3, a transcription factor not DISCUSSION
directly involved in the UPR, were not altered in adipose tissue
of ADGAT1 mice, compared with controls. Additionally, some During lipolysis, adipose tissue FAs are liberated from TG, but a
UPR targets were not changed at the mRNA level under this large fraction of these FAs is re-esterified back to TG (Edens
experimental paradigm. In some instances, changes in mRNA et al., 1990; Leibel et al., 1985; Nye et al., 2008; Vaughan,
levels of UPR genes between ADGAT1 and control adipose tis- 1962). Neither the mechanism for TG synthesis nor the reasons
sue were mirrored by changes in protein expression levels. Spe- for this FA re-esterification cycle have been clear. Here we find
cifically, WAT of 16 hr fasted ADGAT1 mice showed increased that, during lipolysis, DGAT1 is activated and mediates this re-
levels of BIP and CHOP (an effector of the PERK branch of the esterification, whereas DGAT2 is inactivated. Surprisingly, we
UPR), compared with controls (Figures 6B and S6). find that the function of DGAT1-mediated FA re-esterification is
not to preserve TG mass but instead protects the ER from lipo-
Adipocyte-Specific DGAT1 Deficiency Induces the toxic stress and, as shown in mice lacking DGAT1 in adipocytes,
Inflammatory Activation of Adipose Tissue associated adipose tissue inflammation.
Macrophages during Fasting or Cold Exposure More generally, our data indicate that DGAT-mediated TG
Prolonged ER stress in WAT is usually associated with synthesis is important to prevent adipocyte ER stress under
increased inflammation (Hotamisligil, 2010; Kaser et al., 2008; both basal and lipolysis-stimulating conditions. Our findings
Keestra-Gounder et al., 2016; Montane et al., 2014; Urano are consistent with earlier reports that generally suggested
et al., 2000). Thus, we next examined the level of inflammation DGAT1 function is important to avert cellular lipotoxicity. For
in WAT during fasting or in the cold. After a 16 hr fast or a 6 hr example, murine fibroblasts lacking DGAT1 are sensitive to
cold exposure in the absence of food, WAT from ADGAT1 mice oleate-induced cell death (Listenberger et al., 2003). Addition-
showed higher mRNA levels of F4/80 and Cd68, reflecting a ally, several tissue-specific transgenic models of DGAT1 overex-
larger number of activated macrophages in this tissue than in pression have reported beneficial effects that are consistent with
controls. We also observed greater expression of M1, proin- a role of DGAT1-mediated ER protection (Chen et al., 2002b; Ko-
flammatory markers such as Tnf, Il1b, and Ccl2 in WAT from liwad et al., 2010; Liu et al., 2009), and deletion of DGAT1 in heart
fasted or cold exposed ADGAT1 mice than in controls, which increased lipotoxicity in a murine model (Liu et al., 2014). We now
was associated with modest increases in serum cytokines show a mechanism for this protection: the detoxification of lipid
and inflammatory markers (Figures 6C and 6D) In contrast, species by DGAT1 prevents buildup of toxic lipids in the ER that
transcripts reflecting the presence of anti-inflammatory M2 can lead to sustained UPR activation. Although this study
macrophages, such as Il4, Il10, and Arg1 were similar between focused on DGAT1, activity of either DGAT enzyme was suffi-
the genotypes (Figure 6C). cient to protect the adipocyte ER from lipotoxic stress under
basal conditions. We note that our findings do not address the
Inverse Correlation of DGAT1 Expression with ER Stress fed condition, where re-esterification has also been reported,
Genes in Adipose Tissue of Human Subjects and is under the influence of insulin- and acylation-stimulating
To test whether our findings in murine model systems are valid in protein (Van Harmelen et al., 1999).
humans, we differentiated human multipotent adipose-derived The detoxifying function of DGAT1 is consistent with several
stem (hMADS) cells-3 to adipocytes and measured the ER stress features of the enzyme. First, DGAT1 localizes exclusively to
response in basal conditions and with stimulated lipolysis under the ER (Wilfling et al., 2013), where toxic lipids can accumulate.

Cell Metabolism 26, 407–418, August 1, 2017 413


Controls ADGAT1 KO
M1 genes M2 genes
A C
Xbp1t 4 Tnf ** Il1b Il4
2 * 4
4
** *
2
1 2 2

0 0 0 0
** Il10
Xbp1s 8 F4/80 Ccl2
Relative mRNA
*
6
4
4 ** * 2
4
Relative mRNA

*
2 * 2
0 0 0 0
Bip 10 Cd68 ** Nos2 Arg1
** ** 2
3
3
6
2 * * 1 2
1 * 1
2
0 0 0 0
Atf3 ad lib fasted cold ad lib fasted cold ad lib fasted cold
4

D Controls
2 IL6 ** MCP1 **
19
30 * ADGAT1 KO
Serum Level (pg/ml)

0 20
ad lib fasted cold 3 *
10

0 0
B IL10 TNFα *
Bip * 20
15
*
CHOP 10
10
Tubulin 5

Controls ADGAT1 KO 0 0
fasted cold fasted cold

Figure 6. Lack of DGAT1 in Adipose Tissue Increases the Unfolded Protein Response and Associated Inflammation during Fasting or Cold
Exposure
(A) RT-qPCR analysis of ER stress marker genes in WAT of ADGAT1 KO and control mice fed ad libitum, fasted for 16 hr, or 6 hr cold exposed (while fasting).
(B) Western blot analysis of Bip and CHOP in WAT of 16 hr fasted mice.
(C) Relative mRNA levels of inflammatory genes in WAT of ad libitum fed, fasted for 16 hr, or 6 hr cold-exposed (while fasting) ADGAT1 KO and control mice
determined by RT-qPCR.
(D) Plasma inflammatory markers were estimated by ELISA. Data are presented as ±SD (n = 5–6 mice per group). Statistical significance was evaluated by
unpaired two-tailed Student’s t test. *p < 0.05; **p < 0.01. See also Figure S6.

Second, DGAT1 has broad substrate specificity and can esterify orthologs Are1 and Are2, esterify sterols, a reaction that protects
monacylglycerols, retinol, and long-chain alcohols in addition to cells from accumulation of these lipids. For ACAT enzymes,
DAG (Yen et al., 2005; Yen et al., 2008) and so therefore likely cellular function has been clearly linked to preventing ER stress
protects additionally against toxic accumulation of these lipids due to excess sterols (Devries-Seimon et al., 2005). Maintenance
in the ER. Third, DGAT1 has high activity at high substrate con- of the ER lipid composition and fluidity is likely of crucial impor-
centrations (Cases et al., 2001). Finally, DGAT1 belongs to the tance to cell function, so it is perhaps not surprising that en-
membrane-bound O-acyltransferase (M-BOAT) gene family zymes, such as DGATs and ACATs, monitor and defend against
(Yen et al., 2008), in which several other members function to changes to this composition.
prevent toxic buildup of lipids in the ER. For example, acyl We currently do not know how toxic lipids trigger the UPR
CoA:cholesterol acyltransferase (ACAT) enzymes, or the yeast when DGAT1 activity is absent in adipocytes. The possibility

414 Cell Metabolism 26, 407–418, August 1, 2017


A Figure 7. DGAT1 Expression Is Negatively
THBS1
8
DGAT1 DGAT2 Correlated with ER Stress Genes in Adipose
Tissue of Human Subjects
(A) Correlation of DGAT1 or DGAT2 mRNA levels
6
p-value(-log10)

with genes regulated by the unfolded protein


PDIA5
CREB3L2
response (GO 0006986, n = 167). Results are
CTH SEC61B
CALR TPP1 based on previously published transcriptional
4 HYOU1 EP300 CCND1
DGAT2 profiles generated from human WAT derived from
EP300 ATF6 DGAT1 PREB
DNAJC3 SDF2L1 PARP16 SERP2 DAB2IP 26 non-obese and 30 obese women (Arner et al.,
PDIA6 MANF CREB3L4 BFAR HSPB7 HSPA8
2 SEC31A PTPN1 SEC61A1 2012). Subjects were fasted overnight. Highlighted
TOR1B ADD1
BCL2L11 ACADVL genes (blue circles) are significantly correlating (p <
PTPN2
0.01) with DGAT1 or DGAT2.
0 (B) Correlation between DGAT1 and a subset of the
−0.8 −0.4 0.0 0.4 0.8 −0.8 −0.4 0.0 0.4 0.8 significant genes from (A). Non-obese (n = 26) and
obese (n = 30) subjects are depicted as white and
Correlation (r-value)
black circles, respectively. Data are expressed as
log2 microarray signal.
B 6 7 8 9 10 11 5.5 5.5 6.0 6.5 7.0 7.5 8.0 8.5
9.0
THBS1 PDIA5 ATF6
8.5
DGAT1 log2 microarray signal

8.0 In contrast to DGAT1, we found that


DGAT2 appears to be upregulated with
7.5
re-feeding and inactivated during lipol-
7.0 ysis. This is consistent with a primary
9.0 DNAJC3 PDIA6 CREB3L2 function for DGAT2 in mediating TG stor-
8.5 age (Stone et al., 2004). Under basal con-
ditions in adipocytes, DGAT2 is active
8.0
and can play a role in FA re-esterification,
7.5 as suggested previously (Eichmann et al.,
2012) and by our data. The inactivation
7.0
6.0 6.5 7.0 7.5 8.0 9.0 9.5 10.0 10.5 6.0 6.5 7.0 7.5 of DGAT2 during lipolysis suggests that
log 2 microarray signal adipocytes coordinate the regulation of
TG synthesis and lipolysis: when lipases,
such as ATGL and HSL, are activated
that alterations in the ER lipids result in the accumulation of by b-adrenergic signaling during lipolysis (Zechner et al., 2012),
unfolded proteins in the ER seems unlikely, as we did not find ef- DGAT2-mediated TG synthesis is inhibited. The inactivation of
fects on protein secretion in the absence of DGAT1. Instead, our DGAT2 activity at LDs during stimulated lipolysis would serve
data support a model in which excess amounts of FAs liberated to prevent re-esterification from occurring directly at the LD sur-
from lipolysis alter the ER membrane lipids, which in turn activate face where lipolysis is taking place, allowing for FAs to be avail-
the UPR directly. This is consistent with other studies showing a able for export from cells.
direct activation of the UPR by altered membrane lipid changes Although in the current study we focused on the role of DGATs
(Promlek et al., 2011; Volmer et al., 2013). Which lipid species in re-esterification, our data showing increased release of both
mediate the activation of the UPR during lipolysis in the absence glycerol and fatty acids during induced lipolysis in the setting
of DGAT activity is unknown. One possibility is that changes in of DGAT1 inhibition suggest that lipolysis completely hydrolyzes
ER phospholipid composition (e.g., a higher degree of saturated TGs to their glycerol and FA components. This implies that glyc-
FA side chains) trigger activation of the Ire1 and PERK UPR sen- erol 3-phosphate, from glycolysis or glyceroneogenesis, is the
sors (Robblee et al., 2016; Volmer and Ron, 2015; Volmer likely backbone of ER-based FA re-esterification, a hypothesis
et al., 2013). that can be tested in future studies.
UPR induction is a homeostatic response to resolve cellular How DGAT2 is inactivated in adipocytes during lipolysis is un-
ER stress (Volmer and Ron, 2015; Walter and Ron, 2011). At least known. We found that DGAT2 mRNA levels were downregulated
two lines of evidence suggest that this resolution is not achieved in WAT during lipolytic conditions. Additionally, DGAT2 has
in the absence of DGAT1 during lipolysis. First, cells activate multiple potential phosphorylation sites (Yen et al., 2008), sug-
RIDD to remove messages encoding secreted proteins, which gesting post-translational regulation of its stability or activity.
is a sign of unresolved ER stress in vitro (Han et al., 2009; Hollien DGAT2 is a short-lived protein (T1/2 of 30 min) that is degraded
et al., 2009; Maurel et al., 2014), and we found evidence of RIDD by ER-associated protein degradation (Brandt et al., 2016; Choi
activation in our studies. Second, in mice lacking DGAT1 in adi- et al., 2014). Therefore, fast turnover of DGAT2 could also enable
pocytes that were subjected to lipolysis-stimulating conditions, efficient downregulation of its activity.
we found evidence of inflammation, a known consequence of Several findings, from this study and others, indicate that an
prolonged ER stress (Hotamisligil, 2010; Kaser et al., 2008; ER-protective role of DGAT1 activity is likely conserved in hu-
Keestra-Gounder et al., 2016; Montane et al., 2014; Urano mans. In the current study, we found that DGAT1 inhibition re-
et al., 2000). sulted in increased UPR gene expression in cultured hMADS

Cell Metabolism 26, 407–418, August 1, 2017 415


cells. Further, we found a negative correlation of DGAT1 mRNA AUTHOR CONTRIBUTIONS
levels with many UPR genes in adipose tissue of fasted human
C.C., R.V.F., and T.C.W. planned the study and designed the experiments.
subjects. In previous studies, we and others found that homozy-
C.C. performed most of the experiments. N.M. analyzed mRNA expression
gous DGAT1 loss-of-function mutations in humans lead to a se- profiles of human subjects. J.H. performed DGAT activity assays. L.G.D.-R.
vere congenital diarrheal disorder involving enterocyte dysfunc- and C.A.G. helped to maintain mouse colonies. C.C., R.V.F., and T.C.W. wrote
tion, which is triggered by a fat-containing diet (Gluchowski the manuscript.
et al., 2017; Haas et al., 2012; Stephen et al., 2016). Similarly,
DGAT1 inhibition caused dose-related diarrhea in clinical studies ACKNOWLEDGMENTS
of humans (Denison et al., 2014; Meyers et al., 2015). Importantly,
We thank Ryan S. Streeper, Charles A. Harris, Scot Stone, Meghana Rao, and
DGAT2 appears not to be expressed in human intestine (Haas
Claudio J. Villanueva for initial observations that laid the groundwork for this
et al., 2012), creating a situation similar to adipocyte lipolysis, report. We thank Thi Nguyen for help with q RT-PCR analysis, Michel Becuwe
with only DGAT1 active. Thus, DGAT1 might have a particularly for help with image analysis, Romain Christiano for help with data analysis,
important ER-protective role in the human intestine, and absence Jane Lee for technical assistance, and Gary Howard for manuscript editing.
of this protective function might lead to ER toxicity and entero- This work was supported by an ADA mentor-based fellowship grant (7-12-
cyte dysfunction when cells are exposed to high lipid levels MN-18 to C.C. and R.V.F.), R01 DK101579 (to T.C.W. and R.V.F.), RO1
from the diet. Consistent with this model, mice express both DK056084 (to R.V.F.), R01 GM097194 (to T.C.W.), the Mathers foundation
(to T.C.W.), and seed funding from the J. David Gladstone Institute (to
DGAT enzymes in their intestine (Buhman et al., 2002), and
R.V.F.). T.C.W is an investigator of the Howard Hughes Medical Institute. We
DGAT1 knockout mice do not exhibit diarrhea or other overt in- thank Shirly Pinto, Merck & Co., Inc., for gift of DGAT inhibitors, and Dominique
testinal consequences (Buhman et al., 2002; Smith et al., 2000). Langin for gift of the hMADS-3 cells.
In summary, our data provide new insight into the function of
FA re-esterification in WAT during lipolysis. Periods of fasting Received: October 24, 2016
in animals result in mobilization of large amounts of FA from Revised: April 10, 2017
Accepted: July 17, 2017
WAT to provide fuel for other tissues. Re-esterification of a large
Published: August 1, 2017
portion of these FAs was thought to preserve fat mass. Instead,
our findings indicate that DGAT1-mediated re-esterification REFERENCES
plays a crucial role in protecting adipocytes from ER toxicity un-
der such FA-liberating conditions. Protection of the ER by DGAT Ariotti, N., Murphy, S., Hamilton, N.A., Wu, L., Green, K., Schieber, N.L., Li, P.,
enzymes may be important in many tissues routinely challenged Martin, S., and Parton, R.G. (2012). Postlipolytic insulin-dependent remodeling
by huge fluctuations in FA concentrations, such as adipocytes or of micro lipid droplets in adipocytes. Mol. Biol. Cell 23, 1826–1837.

intestine, allowing organisms to adapt to constantly changing Arner, E., Mejhert, N., Kulyté, A., Balwierz, P.J., Pachkov, M., Cormont, M.,
Lorente-Cebrián, S., Ehrlund, A., Laurencikiene, J., Hedén, P., et al. (2012).
energy conditions.
Adipose tissue microRNAs as regulators of CCL2 production in human
obesity. Diabetes 61, 1986–1993.
STAR+METHODS Bezaire, V., Mairal, A., Ribet, C., Lefort, C., Girousse, A., Jocken, J.,
Laurencikiene, J., Anesia, R., Rodriguez, A.M., Ryden, M., et al. (2009).
Detailed methods are provided in the online version of this paper Contribution of adipose triglyceride lipase and hormone-sensitive lipase to
lipolysis in hMADS adipocytes. J. Biol. Chem. 284, 18282–18291.
and include the following:
Brandt, C., McFie, P.J., and Stone, S.J. (2016). Diacylglycerol acyltransfer-
d KEY RESOURCES TABLE ase-2 and monoacylglycerol acyltransferase-2 are ubiquitinated proteins
d CONTACT FOR REAGENT AND RESOURCE SHARING that are degraded by the 26S proteasome. Biochem. J. 473, 3621–3637.

d METHOD DETAILS Buhman, K.K., Smith, S.J., Stone, S.J., Repa, J.J., Wong, J.S., Knapp, F.F., Jr.,
Burri, B.J., Hamilton, R.L., Abumrad, N.A., and Farese, R.V., Jr. (2002). DGAT1
B Animals and Dietary Intervention Studies
is not essential for intestinal triacylglycerol absorption or chylomicron synthe-
B 3T3-L1 Cell Differentiation
sis. J. Biol. Chem. 277, 25474–25479.
B Culture and Differentiation of Human Multipotent Adi-
Cases, S., Smith, S.J., Zheng, Y.W., Myers, H.M., Lear, S.R., Sande, E., Novak,
pose-derived Stem Cells S., Collins, C., Welch, C.B., Lusis, A.J., et al. (1998). Identification of a gene
B [14C]- Oleic Acid Labeling of Lipids, Lipid Extraction encoding an acyl CoA:diacylglycerol acyltransferase, a key enzyme in triacyl-
and Thin Layer Chromatography glycerol synthesis. Proc. Natl. Acad. Sci. USA 95, 13018–13023.
B DGAT Activity Assay Cases, S., Stone, S.J., Zhou, P., Yen, E., Tow, B., Lardizabal, K.D., Voelker, T.,
B Ex Vivo Lipolysis Assay and Farese, R.V., Jr. (2001). Cloning of DGAT2, a second mammalian diacyl-
B Microscopy and Image Processing glycerol acyltransferase, and related family members. J. Biol. Chem. 276,
38870–38876.
B RNA Extraction and Quantitative Real-Time PCR
(qRT-PCR) Chen, H.C., Smith, S.J., Ladha, Z., Jensen, D.R., Ferreira, L.D., Pulawa, L.K.,
McGuire, J.G., Pitas, R.E., Eckel, R.H., and Farese, R.V., Jr. (2002a). Increased
B Immunoblotting
insulin and leptin sensitivity in mice lacking acyl CoA:diacylglycerol acyltrans-
B DGAT Inhibitors
ferase 1. J. Clin. Invest. 109, 1049–1055.
d QUANTIFICATION AND STATISTICAL ANALYSIS
Chen, H.C., Stone, S.J., Zhou, P., Buhman, K.K., and Farese, R.V., Jr. (2002b).
Dissociation of obesity and impaired glucose disposal in mice overexpressing
SUPPLEMENTAL INFORMATION acyl coenzyme a:diacylglycerol acyltransferase 1 in white adipose tissue.
Diabetes 51, 3189–3195.
Supplemental Information includes seven figures and one table and can be Choi, K., Kim, H., Kang, H., Lee, S.Y., Lee, S.J., Back, S.H., Lee, S.H., Kim,
found with this article at http://dx.doi.org/10.1016/j.cmet.2017.07.012. M.S., Lee, J.E., Park, J.Y., et al. (2014). Regulation of diacylglycerol

416 Cell Metabolism 26, 407–418, August 1, 2017


acyltransferase 2 protein stability by gp78-associated endoplasmic-reticulum- C.R., et al. (2016). NOD1 and NOD2 signalling links ER stress with inflamma-
associated degradation. FEBS J. 281, 3048–3060. tion. Nature 532, 394–397.
Denison, H., Nilsson, C., Löfgren, L., Himmelmann, A., Mårtensson, G., Koliwad, S.K., Streeper, R.S., Monetti, M., Cornelissen, I., Chan, L., Terayama,
Knutsson, M., Al-Shurbaji, A., Tornqvist, H., and Eriksson, J.W. (2014). K., Naylor, S., Rao, M., Hubbard, B., and Farese, R.V., Jr. (2010). DGAT1-
Diacylglycerol acyltransferase 1 inhibition with AZD7687 alters lipid handling dependent triacylglycerol storage by macrophages protects mice from diet-
and hormone secretion in the gut with intolerable side effects: a randomized induced insulin resistance and inflammation. J. Clin. Invest. 120, 756–767.
clinical trial. Diabetes Obes. Metab. 16, 334–343. Kuerschner, L., Moessinger, C., and Thiele, C. (2008). Imaging of lipid biosyn-
Devries-Seimon, T., Li, Y., Yao, P.M., Stone, E., Wang, Y., Davis, R.J., Flavell, thesis: how a neutral lipid enters lipid droplets. Traffic 9, 338–352.
R., and Tabas, I. (2005). Cholesterol-induced macrophage apoptosis requires Leibel, R.L., Hirsch, J., Berry, E.M., and Gruen, R.K. (1985). Alterations in
ER stress pathways and engagement of the type A scavenger receptor. J. Cell adipocyte free fatty acid re-esterification associated with obesity and weight
Biol. 171, 61–73. reduction in man. Am. J. Clin. Nutr. 42, 198–206.
Duncan, R.E., Ahmadian, M., Jaworski, K., Sarkadi-Nagy, E., and Sul, H.S. Listenberger, L.L., Han, X., Lewis, S.E., Cases, S., Farese, R.V., Jr., Ory, D.S.,
(2007). Regulation of lipolysis in adipocytes. Annu. Rev. Nutr. 27, 79–101. and Schaffer, J.E. (2003). Triglyceride accumulation protects against fatty
Edens, N.K., Leibel, R.L., and Hirsch, J. (1990). Mechanism of free fatty acid re- acid-induced lipotoxicity. Proc. Natl. Acad. Sci. USA 100, 3077–3082.
esterification in human adipocytes in vitro. J. Lipid Res. 31, 1423–1431. Liu, L., Shi, X., Bharadwaj, K.G., Ikeda, S., Yamashita, H., Yagyu, H., Schaffer,
Eguchi, J., Wang, X., Yu, S., Kershaw, E.E., Chiu, P.C., Dushay, J., Estall, J.L., J.E., Yu, Y.H., and Goldberg, I.J. (2009). DGAT1 expression increases
Klein, U., Maratos-Flier, E., and Rosen, E.D. (2011). Transcriptional control of heart triglyceride content but ameliorates lipotoxicity. J. Biol. Chem. 284,
adipose lipid handling by IRF4. Cell Metab. 13, 249–259. 36312–36323.
Eichmann, T.O., Kumari, M., Haas, J.T., Farese, R.V., Jr., Zimmermann, R., Liu, Q., Siloto, R.M., Lehner, R., Stone, S.J., and Weselake, R.J. (2012).
Lass, A., and Zechner, R. (2012). Studies on the substrate and stereo/regiose- Acyl-CoA:diacylglycerol acyltransferase: molecular biology, biochemistry
lectivity of adipose triglyceride lipase, hormone-sensitive lipase, and diacylgly- and biotechnology. Prog. Lipid Res. 51, 350–377.
cerol-O-acyltransferases. J. Biol. Chem. 287, 41446–41457. Liu, J., Gorski, J.N., Gold, S.J., Chen, D., Chen, S., Forrest, G., Itoh, Y., Marsh,
Farese, R.V., Jr., and Walther, T.C. (2009). Lipid droplets finally get a little D.J., McLaren, D.G., Shen, Z., et al. (2013). Pharmacological inhibition of diac-
R-E-S-P-E-C-T. Cell 139, 855–860. ylglycerol acyltransferase 1 reduces body weight and modulates gut peptide
release–potential insight into mechanism of action. Obesity (Silver Spring)
Fujimoto, T., and Parton, R.G. (2011). Not just fat: the structure and function
21, 1406–1415.
of the lipid droplet. Cold Spring Harb. Perspect. Biol. 3, http://dx.doi.org/10.
1101/cshperspect.a004838. Liu, L., Trent, C.M., Fang, X., Son, N.H., Jiang, H., Blaner, W.S., Hu, Y., Yin,
Y.X., Farese, R.V., Jr., Homma, S., et al. (2014). Cardiomyocyte-specific loss
Gluchowski, N.L., Chitraju, C., Picoraro, J.A., Mejhert, N., Pinto, S., Xin, W.,
of diacylglycerol acyltransferase 1 (DGAT1) reproduces the abnormalities in
Kamin, D.S., Winter, H.S., Chung, W.K., Walther, T.C., and Farese, R.V., Jr.
lipids found in severe heart failure. J. Biol. Chem. 289, 29881–29891.
(2017). Identification and characterization of a novel DGAT1 missense muta-
tion associated with congenital diarrhea. J. Lipid Res. 58, 1230–1237. Marcinkiewicz, A., Gauthier, D., Garcia, A., and Brasaemle, D.L. (2006). The
phosphorylation of serine 492 of perilipin a directs lipid droplet fragmentation
Guan, H.P., Li, Y., Jensen, M.V., Newgard, C.B., Steppan, C.M., and Lazar, and dispersion. J. Biol. Chem. 281, 11901–11909.
M.A. (2002). A futile metabolic cycle activated in adipocytes by antidiabetic
Maurel, M., Chevet, E., Tavernier, J., and Gerlo, S. (2014). Getting RIDD of
agents. Nat. Med. 8, 1122–1128.
RNA: IRE1 in cell fate regulation. Trends Biochem. Sci. 39, 245–254.
Haas, J.T., Winter, H.S., Lim, E., Kirby, A., Blumenstiel, B., DeFelice, M.,
Meyers, C.D., Tremblay, K., Amer, A., Chen, J., Jiang, L., and Gaudet, D.
Gabriel, S., Jalas, C., Branski, D., Grueter, C.A., et al. (2012). DGAT1 mutation
(2015). Effect of the DGAT1 inhibitor pradigastat on triglyceride and apoB48
is linked to a congenital diarrheal disorder. J. Clin. Invest. 122, 4680–4684.
levels in patients with familial chylomicronemia syndrome. Lipids Health Dis.
Han, D., Lerner, A.G., Vande Walle, L., Upton, J.P., Xu, W., Hagen, A., Backes, 14, 8.
B.J., Oakes, S.A., and Papa, F.R. (2009). IRE1alpha kinase activation modes
Montane, J., Cadavez, L., and Novials, A. (2014). Stress and the inflammatory
control alternate endoribonuclease outputs to determine divergent cell fates.
process: a major cause of pancreatic cell death in type 2 diabetes. Diabetes
Cell 138, 562–575.
Metab. Syndr. Obes. 7, 25–34.
Hashimoto, T., Segawa, H., Okuno, M., Kano, H., Hamaguchi, H.O.,
Nye, C., Kim, J., Kalhan, S.C., and Hanson, R.W. (2008). Reassessing triglyc-
Haraguchi, T., Hiraoka, Y., Hasui, S., Yamaguchi, T., Hirose, F., and Osumi,
eride synthesis in adipose tissue. Trends Endocrinol. Metab. 19, 356–361.
T. (2012). Active involvement of micro-lipid droplets and lipid-droplet-associ-
ated proteins in hormone-stimulated lipolysis in adipocytes. J. Cell Sci. 125, Paar, M., Ju€ngst, C., Steiner, N.A., Magnes, C., Sinner, F., Kolb, D., Lass, A.,
6127–6136. Zimmermann, R., Zumbusch, A., Kohlwein, S.D., and Wolinski, H. (2012).
Remodeling of lipid droplets during lipolysis and growth in adipocytes.
Hollien, J., and Weissman, J.S. (2006). Decay of endoplasmic reticulum-local-
J. Biol. Chem. 287, 11164–11173.
ized mRNAs during the unfolded protein response. Science 313, 104–107.
Promlek, T., Ishiwata-Kimata, Y., Shido, M., Sakuramoto, M., Kohno, K., and
Hollien, J., Lin, J.H., Li, H., Stevens, N., Walter, P., and Weissman, J.S. (2009).
Kimata, Y. (2011). Membrane aberrancy and unfolded proteins activate the
Regulated Ire1-dependent decay of messenger RNAs in mammalian cells.
endoplasmic reticulum stress sensor Ire1 in different ways. Mol. Biol. Cell
J. Cell Biol. 186, 323–331.
22, 3520–3532.
Hotamisligil, G.S. (2010). Endoplasmic reticulum stress and the inflammatory Robblee, M.M., Kim, C.C., Porter Abate, J., Valdearcos, M., Sandlund, K.L.,
basis of metabolic disease. Cell 140, 900–917. Shenoy, M.K., Volmer, R., Iwawaki, T., and Koliwad, S.K. (2016). Saturated
Imbriglio, J.E., Shen, D.M., Liang, R., Marby, K., You, M., Youm, H.W., Feng, fatty acids engage an IRE1a-dependent pathway to activate the NLRP3 in-
Z., London, C., Xiong, Y., Tata, J., et al. (2015). Discovery and pharmacology flammasome in myeloid cells. Cell Rep. 14, 2611–2623.
of a novel class of diacylglycerol acyltransferase 2 inhibitors. J. Med. Chem. Smith, S.J., Cases, S., Jensen, D.R., Chen, H.C., Sande, E., Tow, B., Sanan,
58, 9345–9353. D.A., Raber, J., Eckel, R.H., and Farese, R.V., Jr. (2000). Obesity resistance
Kaser, A., Lee, A.H., Franke, A., Glickman, J.N., Zeissig, S., Tilg, H., and multiple mechanisms of triglyceride synthesis in mice lacking Dgat. Nat.
Nieuwenhuis, E.E., Higgins, D.E., Schreiber, S., Glimcher, L.H., and Genet. 25, 87–90.
Blumberg, R.S. (2008). XBP1 links ER stress to intestinal inflammation and Stephen, J., Vilboux, T., Haberman, Y., Pri-Chen, H., Pode-Shakked, B.,
confers genetic risk for human inflammatory bowel disease. Cell 134, 743–756. Mazaheri, S., Marek-Yagel, D., Barel, O., Di Segni, A., Eyal, E., et al. (2016).
Keestra-Gounder, A.M., Byndloss, M.X., Seyffert, N., Young, B.M., Chávez- Congenital protein losing enteropathy: an inborn error of lipid metabolism
Arroyo, A., Tsai, A.Y., Cevallos, S.A., Winter, M.G., Pham, O.H., Tiffany, due to DGAT1 mutations. Eur. J. Hum. Genet. 24, 1268–1273.

Cell Metabolism 26, 407–418, August 1, 2017 417


Stone, S.J., Myers, H.M., Watkins, S.M., Brown, B.E., Feingold, K.R., Elias, through their transmembrane domains. Proc. Natl. Acad. Sci. USA 110,
P.M., and Farese, R.V., Jr. (2004). Lipopenia and skin barrier abnormalities 4628–4633.
in DGAT2-deficient mice. J. Biol. Chem. 279, 11767–11776.
Walter, P., and Ron, D. (2011). The unfolded protein response: from stress
Stone, S.J., Levin, M.C., Zhou, P., Han, J., Walther, T.C., and Farese, R.V., Jr. pathway to homeostatic regulation. Science 334, 1081–1086.
(2009). The endoplasmic reticulum enzyme DGAT2 is found in mitochondria-
associated membranes and has a mitochondrial targeting signal that pro- Walther, T.C., and Farese, R.V., Jr. (2012). Lipid droplets and cellular lipid
motes its association with mitochondria. J. Biol. Chem. 284, 5352–5361. metabolism. Annu. Rev. Biochem. 81, 687–714.
Tordjman, J., Chauvet, G., Quette, J., Beale, E.G., Forest, C., and Antoine, B. Wilfling, F., Wang, H., Haas, J.T., Krahmer, N., Gould, T.J., Uchida, A., Cheng,
(2003). Thiazolidinediones block fatty acid release by inducing glyceroneogen- J.X., Graham, M., Christiano, R., Fröhlich, F., et al. (2013). Triacylglycerol syn-
esis in fat cells. J. Biol. Chem. 278, 18785–18790. thesis enzymes mediate lipid droplet growth by relocalizing from the ER to lipid
Unger, R.H., and Zhou, Y.T. (2001). Lipotoxicity of beta-cells in obesity and in droplets. Dev. Cell 24, 384–399.
other causes of fatty acid spillover. Diabetes 50 (Suppl 1 ), S118–S121. Wood, F.C., Jr., Leboeuf, B., and Cahill, G.F., Jr. (1960). Metabolic role of
Urano, F., Wang, X., Bertolotti, A., Zhang, Y., Chung, P., Harding, H.P., and glucose. A source of glyceride-glycerol in controlling the release of fatty acids
Ron, D. (2000). Coupling of stress in the ER to activation of JNK protein kinases by adipose tissue. Diabetes 9, 261–263.
by transmembrane protein kinase IRE1. Science 287, 664–666.
Yen, C.L., Monetti, M., Burri, B.J., and Farese, R.V., Jr. (2005). The triacylgly-
Van Harmelen, V., Reynisdottir, S., Cianflone, K., Degerman, E., Hoffstedt, J.,
cerol synthesis enzyme DGAT1 also catalyzes the synthesis of diacylglycerols,
Nilsell, K., Sniderman, A., and Arner, P. (1999). Mechanisms involved in the
waxes, and retinyl esters. J. Lipid Res. 46, 1502–1511.
regulation of free fatty acid release from isolated human fat cells by acyla-
tion-stimulating protein and insulin. J. Biol. Chem. 274, 18243–18251. Yen, C.L., Stone, S.J., Koliwad, S., Harris, C., and Farese, R.V., Jr. (2008).
Vaughan, M. (1962). The production and release of glycerol by adipose tissue Thematic review series: glycerolipids. DGAT enzymes and triacylglycerol
incubated in vitro. J. Biol. Chem. 237, 3354–3358. biosynthesis. J. Lipid Res. 49, 2283–2301.

Villanueva, C.J., Monetti, M., Shih, M., Zhou, P., Watkins, S.M., Bhanot, S., and Zechner, R., Zimmermann, R., Eichmann, T.O., Kohlwein, S.D., Haemmerle,
Farese, R.V., Jr. (2009). Specific role for acyl CoA:Diacylglycerol acyltransfer- G., Lass, A., and Madeo, F. (2012). FAT SIGNALS—lipases and lipolysis in lipid
ase 1 (Dgat1) in hepatic steatosis due to exogenous fatty acids. Hepatology metabolism and signaling. Cell Metab. 15, 279–291.
50, 434–442.
Zhao, T.J., Liang, G., Li, R.L., Xie, X., Sleeman, M.W., Murphy, A.J.,
Volmer, R., and Ron, D. (2015). Lipid-dependent regulation of the unfolded Valenzuela, D.M., Yancopoulos, G.D., Goldstein, J.L., and Brown, M.S.
protein response. Curr. Opin. Cell Biol. 33, 67–73. (2010). Ghrelin O-acyltransferase (GOAT) is essential for growth hormone-
Volmer, R., van der Ploeg, K., and Ron, D. (2013). Membrane lipid saturation mediated survival of calorie-restricted mice. Proc. Natl. Acad. Sci. USA 107,
activates endoplasmic reticulum unfolded protein response transducers 7467–7472.

418 Cell Metabolism 26, 407–418, August 1, 2017


STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER


Antibodies
BiP (C50B12) Rabbit mAb Cell Signaling Technology Cat # 3177
CHOP (L63F7) Mouse mAb Cell Signaling Technology Cat # 2895
Perilipin Rabbit mAb Cell Signaling Technology Cat # 9349
ATGL Rabbit mAb Cell Signaling Technology Cat # 2439
HSL Antibody Cell Signaling Technology Cat # 4107
Phospho-HSL (Ser660) Antibody Cell Signaling Technology Cat # 4126
DGAT1 Custom made N/A
Anti-a-Tubulin Sigma-Aldrich Cat # T9026
Chemicals, Peptides and Recombinant Proteins
Power SYBR Green PCR Master Mix Life Technologies Cat # 4368706
iScript cDNA Synthesis Kit Biorad Cat # 170-8891
RNeasy Mini Kit Qiagen Cat # 74106
QIAzol Lysis Reagent Qiagen Cat # 79306
Isobutylmethylxantine Sigma-Aldrich Cat # I7018
Dexamethasone Sigma-Aldrich Cat # D4902
Bovine insulin Sigma-Aldrich Cat # I0516
Human insulin Sigma-Aldrich Cat # I 9278
FGF2 human Sigma-Aldrich Cat # F 0291
Transferrine Sigma-Aldrich Cat # T 8158
0
T3 (3,3 ,5-Triiodo-L-thyronine sodium salt) Sigma-Aldrich Cat # T 6397
Rosiglitazone Cayman Chemicals Cat # 71740
BODIPY (493/503) Thermo Fisher Cat # D3922
Isoprenaline hydrochloride Sigma-Aldrich Cat # I5627
Forskolin Sigma-Aldrich Cat # F6886
1,2-Dioleoyl-rac-glycerol Sigma-Aldrich Cat # D8394
Oleoyl coenzyme A lithium salt Sigma-Aldrich Cat # O1012
Oleoyl [1-14C] Coenzyme A American Radiolabeled Chemicals, Inc. ARC 0527
Oleic acid [1-14C] American Radiolabeled Chemicals, Inc. ARC 0297
Thin Layer Chromatography Plates Analtech P43911
DGAT1 inhibitor Merck & Co., Inc. Liu et al., 2013
DGAT2 inhibitor Merck & Co., Inc. Imbriglio et al., 2015
ATGL inhibitor (Atglistatin) Cayman Chemicals Cat # 15284
HSL inhibitor Cayman Chemicals Cat # CAY10499
Triacsin C Santa Cruz Cat # sc-200574A
Thapsigargin Cayman Chemicals Cat # 10522
Tunicamycin Cayman Chemicals Cat # 11445
Critical Commercial Assays
LEGEND MAX Mouse TNF-a ELISA Kit BioLegend Cat # 430907
LEGEND MAX Mouse MCP-1/CCL2 ELISA Kit BioLegend Cat # 432707
LEGEND MAX Mouse IL-6 ELISA Kit BioLegend Cat # 431307
LEGEND MAX Mouse IL-10 ELISA Kit BioLegend Cat # 431417
Infinity Triglycerides Reagent Thermo Fisher Cat # TR22421
Free Glycerol Reagent Sigma-Aldrich Cat # F6428

Cell Metabolism 26, 407–418.e1–e3, August 1, 2017 e1


CONTACT FOR REAGENT AND RESOURCE SHARING

Further information and request for reagent and resources should be directed to Robert V. Farese, Jr. (robert@hsph.harvard.edu) or
Tobias C. Walther (twalther@hsph.harvard.edu).

METHOD DETAILS

Animals and Dietary Intervention Studies


All animal experiments were performed under the guidelines established by Harvard Center for Comparative Medicine (HCCM). Mice
were maintained in a barrier facility, at normal room temperatures, on a regular 12-h light and 12-h dark cycle and had ad libitum
access to food and water unless otherwise stated. ADGAT1 KO mice were generated from DGAT1 floxed mice (Villanueva et al.,
2009) that were crossed with adiponectin Cre mice (a gift from Dr. E. Rosen, (Eguchi et al., 2011)). The genetic background of
mice in this study was C57BL/6J (Jackson Laboratory Bar Harbor, ME). Mice were fed on standard laboratory chow diet (less
than 4.5% crude fat). Calorie restriction study was carried out as described (Zhao et al., 2010). Mice were individually housed for
a week and fed chow diet ad libitum. During this week of acclimation, we monitored the daily food intake of each mouse. The following
week onward, mice were subjected to 60% calorie restriction such that each mouse was fed every day at 6 pm with an amount of food
equal to 40% of their daily intake by the same mouse during the week of acclimation. Body weights were measured daily at 5:30 pm
before feeding: lean and fat mass were determined every 5 days by DEXA.

3T3-L1 Cell Differentiation


3T3-L1 cells were cultured in high glucose DMEM containing 10% FBS and penicillin and streptomycin. Two days after full conflu-
ency cells were cultured for 3 days in differentiation medium containing 1mM dexamethasone, 100 mM isobutylmethylxantine and
5 mg/ml bovine insulin. Cells were then cultured in post differentiation medium containing 5 mg/ml bovine insulin for five to eight
days before using them for metabolic labeling experiments. For confocal microscopy cells were cultured in gelatin coated glass
bottom dishes.

Culture and Differentiation of Human Multipotent Adipose-derived Stem Cells


hMADS-3 cells (a gift from Dominique Langin) were cultured and differentiated as described (Bezaire et al., 2009). Cells were cultured
in proliferation medium (DMEM-low glucose containing 10% FBS, L-glutamine, HEPES Buffer, and 5 mg/ml of FGF2. Proliferation
medium (+FGF2) was changed 3 days after cell plating, and then changed to proliferation medium without FGF2. 6 days after plating.
Then cells were washed with 1X PBS for two times and differentiation medium (proliferation medium without serum/ Ham’s F12 con-
taining insulin, 5 mg/ml; transferrin, 10 mg/ml; T3, 0.2 nM; rosiglitazone, 0.1 mM; IBMX, 100 mM; dexamethasone, 1mM) was added.
Cells were cultured for 3 days. Then cells were washed with 1X PBS for two times and differentiation medium without IBMX and dexa-
methasone was added and incubated for 2-3 days followed by differentiation medium without rosiglitazone. At day 18 of culture, cells
were utilized for experiments.

[14C]- Oleic Acid Labeling of Lipids, Lipid Extraction and Thin Layer Chromatography
DGAT1 and DGAT2 specific activities in differentiated 3T3-L1 adipocytes were determined by [14C]-oleic acid incorporation into TG
in presence of inhibitors. Cells were pre-treated for 30 min with 5 mM inhibitors, cells were pulse labeled with [14C]-oleic acid
(50 mCi/mmol) for 3 hr in presence of inhibitors. To determine activities during induced lipolysis, cells were treated with 10 mM isopro-
terenol. Cells were washed with phosphate buffer saline for 3 times. Lipids were extracted directly from 6-well cell-culture plates by
adding hexane: isopropanol mixture (3:2) and gentle shaking for 10 min. The process is repeated second time for efficient extraction
of all lipids. Lipids were dried under nitrogen stream and separated by TLC using hexane:diethyl ether:acetic acid (80:20:1) solvent
system. TLC plate were exposed to phosphor imaging cassette overnight and revealed by Typhoon FLA 7000 phosphor imager.
Lipids on TLC plate were stained with iodine vapors; bands were scraped and quantified by liquid scintillation counter. After lipid
extraction from 6-well plates, 400 mL of lysis buffer (0.3 N NaOH and 0.1% SDS) was added to each well and kept for shaking for
3 hr to lyse the cells for protein measurement by Bio-Rad DC Protein Assay kit.

DGAT Activity Assay


DGAT1- and DGAT2-specific enzymatic activities were measured in membrane fractions isolated from WAT of wild-type and DGAT1
KO mice, respectively. Enzymatic activities were measured at Vmax substrate concentrations. Assay mixture contained 5–10 mg of
membrane proteins, 100 mM of 1,2-dioleoyl-sn-glycerol, 25 mM of oleoyl-CoA, which contained [14C] oleoyl-CoA as tracer, 25 mM
MgCl2 for the DGAT1 assay and 1 mM MgCl2 for the DGAT2 assay in an assay in buffer containing 100 mM Tris-HCl (pH 7.4) and
protease inhibitors. Reaction was carried out as described (Cases et al., 2001; Yen et al., 2005). After stopping the reaction, lipids
were extracted, then separated by TLC using hexane:diethyl ether:acetic acid (80:20:1) solvent system. The TLC plates were
exposed to phosphor imager screen and developed.

e2 Cell Metabolism 26, 407–418.e1–e3, August 1, 2017


Ex Vivo Lipolysis Assay
Mice which had ad libitum access to food and water were sacrificed and WAT was isolated and cut in to pieces in phosphate buffer
saline (PBS). To measure basal lipolysis, small pieces (2–3 mm) of WAT were incubated in 96-well plate in serum-free medium con-
taining 1% fatty acid–free BSA. To measure glycerol and fatty acid release during stimulated lipolysis, WAT explants were incubated
in were incubated in 96-well plate in serum-free medium containing 1% fatty acid–free BSA and 10 mM isoproterenol. Glycerol release
into medium was measured using free glycerol reagent kit from Sigma, fatty acid release was measured using NEFA kit from Wako
diagnostics. according to manufacturer’s protocol.

Microscopy and Image Processing


Microscopy was performed on spinning disk confocal (Yokogawa CSU-X1) set up on a Nikon Eclipse Ti inverted microscope with a
100 3 ApoTIRF 1.4 NA objective (Nikon) in line with 2x amplification. BODIPY 493/503 fluorophore was exited on 561-nm laser line,
and fluorescence was detected by an iXon Ultra 897 EMCCD camera (Andor). Acquired images were processed using FIJI software
(http://fiji.sc/). LDs were analyzed using IMARIS software (Bitplane).

RNA Extraction and Quantitative Real-Time PCR (qRT-PCR)


Total RNA from 3T3-L1 cells was isolated using the RNeasy Kit (QIAGEN), according to the manufacturer’s instructions. For isolating
RNA from WAT, RNeasy Lipid Tissue (QIAGEN) was used. Complementary DNA was synthesized using iScript cDNA Synthesis Kit
(Bio-Rad), and qPCR was performed in triplicates using SYBR Green PCR Master Mix Kit (Applied Biosystems). Sequences of the
qPCR primers used are listed in Table S1)

Immunoblotting
Cells were lysed using RIPA lysis buffer. Tissues were lysed in buffer containing 250 mM sucrose, 100 mM Tris-HCl (pH 7.4), and
protease inhibitors in a dounce homogenizer. Proteins were denatured in Laemmli buffer and were separated on 10% SDS-PAGE
gel and transferred to PVDF membrane (Bio-Rad). The membranes were blocked with blocking buffer for 2 hr in TBST containing
5% BSA or 5% milk, and then incubated with primary antibodies overnight. The membranes were then washed with TBST for
10 min x 3 times, and incubated in mouse secondary antibodies (Santa Cruz Biotechnology) at 1:5000 dilutions in blocking buffer.
Membranes was washed again with TBST for 10 min x 3 times and revealed using the Super Signal West Pico kit (Thermo Scientific).
DGAT1 antibodies were a gift from Dr. Jin Ye from UT Southwestern Medical Center. CHOP and Bip antibodies were purchased from
Cell Signaling Technology.

DGAT Inhibitors
DGAT2 and DGAT1 inhibitors were obtained from collaborators at Merck & Co., Inc. DGAT2 and DGAT1 inhibitors are well charac-
terized and published. (Imbriglio et al., 2015; Liu et al., 2013). Inhibitors were dissolved in DMSO. For [14C]-oleic acid labeling studies,
Cells were pre-treated for 30 min with 5 mM inhibitors, cells were pulse labeled with [14C]-oleic acid (50 mCi/mmol) for 3 hr in presence
of inhibitors.

QUANTIFICATION AND STATISTICAL ANALYSIS

Data are presented as means ± SD. Statistical significance was evaluated by unpaired two-tailed Student’s t test using Microsoft
Excel software.

Cell Metabolism 26, 407–418.e1–e3, August 1, 2017 e3

Вам также может понравиться