B3 Niacin Energy Mitochondria

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

THE

JOURNAL • RESEARCH • www.fasebj.org

Niacin fine-tunes energy homeostasis through


canonical GPR109A signaling
Lingyan Ye,1 Zheng Cao,1 Xiangru Lai, Weiwei Wang, Zhiqiang Guo, Lili Yan, Yuyan Wang, Ying Shi,2
and Naiming Zhou3
Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, China

ABSTRACT: The incidence of overweight and obesity has become a global public health problem, constituting a major
risk factor for numerous comorbidities. Despite tremendous efforts, effective pharmacological agents for the
treatment of obesity are still limited. Here, we showed that in contrast to lactate receptor GPR81, niacin receptor
GPR109A-deficient mice had progressive weight gain and hepatic fat accumulation. Using high-fat diet–induced
mouse model of obesity, we demonstrated that niacin treatment apparently protected against obesity without
affecting food intake in wild-type mice but not in GPR109A-deficient mice. Further investigation showed that
niacin treatment led to a remarkable inhibition of hepatic de novo lipogenesis. Additionally, we demonstrated that
niacin treatment triggered brown adipose tissue and/or white adipose tissue thermogenic activity via activation of
GPR109A. Moreover, we observed that mice exposed to niacin exhibited a dramatic decrease in intestinal ab-
sorption of sterols and fatty acids. Taken together, our findings demonstrate that acting on GPR109A, niacin shows
the potential to maintain energy homeostasis through multipathways, representing a potential approach to the
treatment of obesity, diabetes and cardiovascular disease.—Ye, L., Cao, Z., Lai, X., Wang, W., Guo, Z., Yan, L., Wang,
Y., Shi, Y., Zhou, N. Niacin fine-tunes energy homeostasis through canonical GPR109A signaling. FASEB J.
33, 4765–4779 (2019). www.fasebj.org
KEY WORDS: obesity • de novo lipogenesis • thermogenesis • intestinal fat absorption

The incidence of overweight and obesity has become intake and energy expenditure (3, 4). Obesity is considered
a global epidemic, with over 1.9 billion overweight and a major risk factor for numerous comorbidities including
600 million obese individuals worldwide (1, 2). Obesity is major diseases such as cardiovascular disease, diabetes,
caused by an abnormal excess storage of energy as lipids in and cancer (5, 6). However, despite tremendous efforts,
adipose tissue due to a net imbalance between energy effective antiobesity pharmacotherapies are still lim-
ited. Additionally, to date, current antiobesity agents are
designed to reduce food intake and appetite through
ABBREVIATIONS: ACC, acetyl CoA carboxylase; ACC1, acetyl CoA car-
boxylase 1; AUC, area under the curve; BAT, brown adipose tissue; the hypothalamus-based molecular pathways, but un-
DGAT2, diacylglycerol acyltransferase 2; DIO, diet-induced obesity; fortunately, these agents exhibit severe psychiatric or
eWAT, epididymal WAT; FAS, fatty acid synthase; FATP4, fatty acid cardiovascular side effects (7–9). Therefore, the magnitude
transport protein 4; FFA, free fatty acid; GTT, glucose tolerance test; H&E,
hematoxylin and eosin; HFD, high-fat diet; HOMA-IR, homeostasis model
of this current health epidemic has heightened the need
assessment of insulin resistance; I-FABP, intestinal-type fatty acid–binding for alternative therapeutic strategies aiming to control
protein; ITT, insulin tolerance test; NCD, normal chow diet; NPC1L1, body weight through potential targets in peripheral non-
Niemann-Pick C1-Like 1; PDH, pyruvate dehydrogenase; PGC-1a, per- neuronal tissues.
oxisome proliferator-activated receptor g coactivator 1-a; PRDM16, PR
domain containing 16; pWAT, perirenal WAT; qRT-PCR, quantitative Since the discovery of decreasing plasma cholesterol
RT-PCR; SCD-1, stearoyl-CoA desaturase-1; SREBP-1c, sterol regulatory levels in 1955, nicotinic acid (niacin) has been used to treat
element–binding protein-1c; sWAT, subcutaneous WAT; TC, total cho- dyslipidemia as an approved drug for more than half a
lesterol; Tg, triglyceride; UCP1, uncoupling protein 1; WAT, white adi-
pose tissue; WT, wild-type
century (10, 11). A number of randomized clinical trials
1
demonstrated that niacin monotherapy has been shown to
These authors contributed equally to this work.
2
Correspondence: Institute of Biochemistry, College of Life Sciences,
substantially increase levels of HDL-C and decrease levels
Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, of triglycerides (Tgs) (12, 13). However, its exact mecha-
3
China. E-mail: shi_y@zju.edu.cn nism was not well understood until the orphan receptor
Correspondence: Institute of Biochemistry, College of Life Sciences, GPR109A (recently renamed as hydroxyl-carboxylic acid
Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058,
China. E-mail: zhounaiming@zju.edu.cn receptor 2 or HCA2) was identified as a receptor with
high affinity for niacin in 2003, independently by 3 research
doi: 10.1096/fj.201801951R
This article includes supplemental data. Please visit http://www.fasebj.org to groups (14–17). Subsequently, the ketone body b-hydroxy-
obtain this information. butyrate was identified as an endogenous ligand for

0892-6638/19/0033-4765 © FASEB 4765


GPR109A (18). In adipocytes, upon stimulation by niacin, Laboratory Animal Center) in the control group. After 6 wk on an
GPR109A functions via Gi/o proteins to lower intra- HFD, mice were divided randomly into 2 groups. One group
cellular cAMP production, leading to decreased PKA- received niacin (50 mM) (MilliporeSigma, Burlington, MA, USA)
dissolved in drinking water, and the other received vehicle
mediated activation of hormone-sensitive lipase, which in (water) for 8–9 wk as a control. Groups of age- and weight-
turn reduces Tg hydrolysis to free fatty acids (FFAs) (19). matched animals were used in all experiments. Food consump-
This FFA hypothesis is supported by evidence that niacin tion and body weight were recorded weekly. After the modeling
failed to lower FFA and Tgs in GPR109A-knockout mice period, mice were briefly starved overnight (12 h) and anes-
(15). thetized with 3% isoflurane in a dedicated chamber, and the
The established clinical benefits of niacin treatment weights of major organs including fat pads and liver were
on cardiovascular events have been challenged by re- recorded. Whole blood was collected and processed to isolate
serum, and tissues were flash frozen in liquid nitrogen and stored
cent studies showing that niacin exerts its beneficial effects at 280°C for biochemical and histologic analyses. All animal
on GPR109A-mediated antiatherosclerotic activity inde- procedures were conducted in compliance with protocols ap-
pendently from its antilipolytic properties (20, 21). How- proved by the Institutional Animal Care and Use Committee at
ever, these data did not exclude the possibility that niacin Zhejiang University (ZJU2010-1-01-020).
provides cardiovascular benefits in an antilipolysis-
independent manner through GPR109A. A niacin-
Mice primary hepatocyte extraction and culture
mediated beneficial cardiovascular effect has been shown
to be transferable with GPR109A-competent bone mar- Mice (6 wk old) were anesthetized with isoflurane. The liver was
row cells (21). In addition, accumulating evidence has perfused with prewarmed calcium- and magnesium-free HBSS
demonstrated that activation of GPR109A was able to containing 1% penicillin-streptomycin solution, 0.5 mM EDTA,
induce anti-inflammatory effects not only on atheroscle- and 25 mM HEPES at a flow rate of 3.5 ml/min for 5 min until the
rosis but also on acute ischemic stroke, arthritis, chronic liver become pale in color with no blood inside. We then switched
renal failure, and sepsis through inhibition of immune cell to prewarmed digestion buffer [HBSS with calcium-magnesium
containing 1% penicillin-streptomycin solution, 25 mM HEPES,
chemotaxis and proinflammatory cytokine production
5 mM CaCl2, and 0.35 mg/ml collagenase X (Fujifilm, Tokyo,
(20–29). Therefore, more studies are needed to thoroughly Japan)] for 5 min at a flow rate of 3.5 ml/min. We separated the
evaluate the GPR109A-mediated pleiotropic effects on liver with 2 sets of forceps and put the dish into the incubator to
atherogenesis and other metabolic diseases. digest for another 5 min. We then added 25 ml cold DMEM with
GPR109A-induced inhibition in adipocyte lipolysis and 10% FBS and passed the liver through a 100 mm strainer (352360;
fatty acid mobilization in response to long-term treat- Thermo Fisher Scientific, Waltham, MA USA) and isolated a
ment with niacin would result in excessive synthesis of Tgs sample pellet by centrifugation at 50 g for 3 min at 4°C. The liver
was subsequently washed 2 times in 25 ml washing buffer
and obesity (30). However, during breeding process of (DMEM with 1% penicillin-streptomycin solution) and a sample
GPR109A-knockout mice we obtained from Dr. Stefan pellet was isolated by centrifugation at 50 g for 3 min at 4°C. The
Offermanns (Max Planck Institute for Heart and Lung Re- viability of isolated hepatocytes was determined by trypan blue
search, Bad Nauheim, Germany), we found that the exclusion. Hepatocytes were plated in collagen I-coated 6 well
GPR109A-knockout mice are more prone to being obese (5 3 105 cells/well) and 12 well (2 3 105 cells/well) plates in
compared to the wild-type (WT) mice. Therefore, we initi- William E medium (BE02-019; Lonza Group, Basel, Switzerland)
ated this study to investigate roles of niacin-GPR109A in the with 10% FBS and 1% penicillin-streptomycin solution and
then were incubated at 37°C in an atmosphere of 5% CO2. At 2 h
regulation of lipid metabolism using GPR109A-knockout after plating, the plating medium was replaced with fresh culture
mice combined with high-fat diet (HFD)-induced obe- medium consisting of DMEM supplemented with 1.5% FBS,
sity mouse model. We show that upon activation by 1 mM sodium pyruvate, and 1% penicillin-streptomycin solution.
niacin, GPR109A fine tunes lipid metabolism through
multipathways including inhibition of hepatocyte lipo-
Body temperature in cold response
genesis and fatty acid absorption and promotion of brown
adipose tissue (BAT) thermogenesis.
To test resistance to cold exposure, mice were individually caged
and exposed to 4°C for 6 h with free access to water. Anal tem-
perature was monitored at the beginning and hourly after the
MATERIALS AND METHODS start of cold exposure.

Animal studies
Serum collection and parameters determination
All mice were housed in groups of 4–6 per cage under constant
temperature (23–25°C) with a 12-h light/dark cycle. Animals Blood was collected from the orbital sinus, and samples were
were given ad libitum access to water and food. GPR109A het- incubated at room temperature for 30 min to allow clotting. Se-
erozygous mice on C57BL/6 background were a generous gift rum samples were obtained after centrifuging at 3500 rpm at 4°C
from Dr. Stefan Offermanns. Homozygous GPR109A knockout for 20 min. Finally the supernatant (serum) was collected and
and their littermate WT mice were generated by matting het- stored at 280°C. Serum total Tg, total cholesterol (TC), HDL, and
erozygous mice and genotyped using PCR. Male mice were used LDL were measured by using biochemical testing kits (Roche
for all the experiments described in this study. 4-wk-old male Holding AG, Basel, Switzerland). Serum glucose levels were
C57BL/6 mice were purchased from the Shanghai Laboratory measured using commercial kits (Jiancheng, Nanjing, China).
Animal Center (Shanghai, China), and after a week of adaptation, Serum insulin levels were determined using a rat/mouse insulin
mice were fed a HFD (60% kcal from fat) (D12492; Research Diets, ELISA kit (MilliporeSigma) according to the manufacturer’s
New Brunswick, NJ, USA) for generating a diet-induced obesity instructions. Homeostasis model assessment of insulin resis-
(DIO) model and a normal chow diet (NCD) (10% kcal; Shanghai tance (HOMA-IR) was calculated using the following equation:

4766 Vol. 33 April 2019 The FASEB Journal x www.fasebj.org YE ET AL.


HOMA-IR = [fasting plasma insulin (mIU/L)] 3 fasting plasma PCR was performed in 20 ml reaction mixture according to the
glucose (mM)/22.5. instruction of KOD-Plus (Toyobo, Osaka, Japan). Primers used
for GPR109A are indicated in Supplemental Table S1. PCR was
performed using an initial denaturation at 94°C for 5 min, fol-
Fecal and liver lipids analysis lowed by 28 cycles at 94°C for 30 s, 53°C for 30 s, and 68°C for 30 s.
After 28 cycles, an additional elongation step was performed at
Fecal lipids were extracted as previously described (31). Feces 68°C for 10 min. Amplified PCR products were run on 1.2%
were collected from mice housed individually for 5 consecutive agarose gels containing ethidium bromide.
days. Feces were dried for 1 h at 70°C, and 100 mg aliquots of
feces were weighed, incubated with 2 ml of chloroform-methanol
(2:1,v/v) for 30 min at 60°C with constant agitation, and then Histologic analysis
centrifuged (5 min at 5000 rpm). Water (1 ml) was added to the
supernatant; following vortex, phase separation was induced To examine hepatic steatosis, liver samples were embedded in
by low-speed centrifugation (2000 rpm for 10 min). The lower Tissue-Tek (Sakura Finetek, Torrance, CA, USA) and frozen and
chloroform phase was then removed and transferred to a new sectioned (10 mm) and stored at 280°C until use. The sections
tube, and the sample was dried under nitrogen. Then samples were stained with hematoxylin and Oil red O (MilliporeSigma)
were resuspended in 500 ml chloroform, evaporated to dryness, for lipid deposition. For histology, adipose tissues were fixed in
and finally resuspended in 500 ml 100% ethanol. For analysis of 10% neutral-buffered formalin, followed by dehydration with
liver lipids, 100 mg aliquots of tissue were homogenized with graded ethanol (80–100%) and embedding in paraffin. Then the
2 ml chloroform-methanol and then agitated overnight on an or- tissues were sliced into 6-mm pieces using microtome, depar-
bital shaker at 4°C. The homogenate was then centrifuged (5 min at affinized in xylene, and passed through 80–100% ethanol. And
5000 rpm), 0.9% NaCl solution was added to the liquid phase, and hematoxylin and eosin (H&E) staining was performed using
the samples were vortexed. Phase separation was induced by the standard techniques. For transmission electron microscopy
centrifugation (2000 rpm for 10 min), and the bottom phase was analysis, liver and muscle tissues were cut into 3 mm3 fragments
moved to a new tube and then processed as previously described. and fixed by immersion in 2.5% glutaraldehyde in phosphate
The levels of TC, Tg, and FFA in the fecal and liver lipid extracts buffer (0.1 M, pH 7.4) overnight at 4°C. After that, they were
were measured using commercial kits following the manufac- washed in 0.1 M phosphate buffer 3 times. Then, they were
turer’s instructions and normalized to feces or tissue weights. postfixated with 1% osmium tetroxide (OsO4) in 0.1 M phos-
phate buffer for 2 h at room temperature. Subsequently, they
were rinsed with 0.1 M phosphate buffer for 3 times again. Tis-
Glucose and insulin tolerance tests sues were dehydrated through graded alcohols (30, 50, 70, 80, 90,
95, and 100%) for 30 min. Last they were embedded in Epon 812.
For glucose tolerance tests (GTTs), mice were tested after an Ultrathin sections (70–90 nm in thickness) of Epon-embedded
overnight period having food withheld. Glucose (40% solution, samples were placed on copper grids and stained with uranyl
1.5 g/kg body weight; MilliporeSigma) was intraperitoneally acetate and lead citrate. Sections were examined with a Hitachi
injected, and tail vein blood glucose levels were measured at 0, 30, model H-7650 electron microscope (Hitachi, Chiyoda, Japan).
60, 90, and 120 min after injection with a OneTouch Ultra Blood
Glucose Monitoring System (LifeScan, Milpitas, CA, USA). For
insulin tolerance tests (ITTs), mice were briefly starved for 4 h and Measurement of enzymes activities
then intraperitoneally injected with human insulin (0.75 U/kg
body weight; NovoRapid; Novo Nordisk, Bagsværd, Denmark). To determine the activities of enzymes involving in lipogenesis in
Glucose concentrations in blood were measured after 0, 30, 60, 90, liver and white adipose tissue (WAT), the enzyme source fraction
and 120 min. Area under the curves (AUCs) for GTT and ITT was prepared as follows. Briefly, a 10% (w/v) homogenate was
was calculated by trapezoidal approximation, using Prism 6.0 prepared in phosphate buffered solution (pH 7.2–7.4) and then
(GraphPad, La Jolla, CA, USA). centrifuged at 2000–3000 rpm for 20 min and the supernatant was
transferred into a new tube. Samples were stored at 280°C until
use. All enzymes, such as pyruvate dehydrogenase (PDH), acetyl
RNA isolation and quantitative RT-PCR analysis CoA carboxylase (ACC), acetyl CoA carboxylase 1 (ACC1), and
diacylglycerol acyltransferase 2 (DGAT2), were measure by
Total RNA was extracted by using an RNAiso reagent kit ELISA (Jing Ma, Shanghai, China).
(Takara, Otsu, Shiga, Japan). Reverse transcription was per-
formed on each RNA sample (1 mg) by using PrimeScript RT
Reagent Kit with gDNA eraser (Takara) with a final reaction Western blot analysis
volume of 20 ml. For quantitative RT-PCR (qRT-PCR) analysis,
the reaction mixture was run in an iQ5 real time PCR machine Proteins were extracted from tissues or cultured cells by ho-
(Bio-Rad, Hercules, CA, USA) instrument using SYBR Premix mogenizing or scraping in ice-cold RIPA buffer [50 mM Tris (pH
Ex Taq II (Takara). All expression levels were normalized to 7.4), 150 mM NaCl, 1% Triton X-100, 1% sodium deoxycholate,
the corresponding glyceraldehyde-3-phosphate dehydrogenase 0.1% SDS, 2 mM EDTA] (Beyotime, Shanghai, China) containing
mRNA levels and analyzed using the 22DDCt method. The qRT- 1 mM sodium orthovanadate and protease inhibitors (Complete
PCR cycles were as follows: step 1, preparative denaturation (30 s Mini; Roche). Tissue samples were transferred to microcentrifuge
at 95°C); step 2, 40 cycles of denaturation (5 s at 95°C) and tubes and rotated on a rocking platform for 40 min at 4°C after
annealing (30 s at 60°C); and step 3, dissociation following the homogenization on ice. The homogenate was cleared by centri-
manufacturer’s protocol. PCR primers used in the real-time PCR fugation at 4°C for 20 min at 12,000 rpm and the supernatant was
analysis are listed in Supplemental Table S1. transferred into a new tube. Protein concentration in the super-
natant was determined using the BCA Protein Assay Kit (Probe
Gene, Jiangsu, China). Equal amounts of protein samples were
Semiquantification PCR subjected to SDS-PAGE and transferred to PVDF membranes
(MilliporeSigma). Membranes were blocked with 5% bovine se-
The total RNA was extracted by using an RNAiso Reagent Kit. rum albumin in tris-buffered saline containing 0.1% Tween-20
The cDNA were synthesized using PrimeScript RT Reagent Kit. (TBST) for 1 h at room temperature and incubated overnight at

GPR109A REGULATES ENERGY HOMEOSTASIS 4767


4°C with primary antibodies against GPR109A (Santa Cruz with niacin exhibited a significant reduction in adipose
Biotechnology, Dallas, TX, USA) and b-tubulin (Cell Signaling tissue weights (including eWAT, pWAT, and inguinal
Technology, Danvers, MA, USA), respectively. The following WAT) and liver weight, but not in BAT or other organ
day, the membranes were washed with TBST and incubated
with a horseradish peroxidase–conjugated secondary antibody
weights (kidney, lung, heart, and spleen) with respect to
(1: 2000) for 2 h at room temperature. Proteins were visualized vehicle-treated mice (Fig. 2E, F and Supplemental Fig. S1C,
with an ECL reagent (Beyotime) by the Tanon 5200 Chemilu- D), which indicated that niacin protected the mice from
minescent Imaging System (Tanon, Shanghai, China). DIO through GPR109A. Furthermore, histologic exami-
nation showed that niacin treatment led to a marked de-
crease in lipid accumulation in liver, muscle, and eWAT
Statistical analysis
(Fig. 2G).
Statistical analysis was performed using Prism 6.0 (GraphPad). Niacin has profound and unique beneficial effects on
All of the data were expressed as the mean 6 SEM. All experi- blood lipid (11). We examined the effects of niacin treat-
ments were performed in triplicate. Unpaired, 2-tailed Student’s ment on metabolic parameters in serum of experimental
t test was used for comparisons between 2 groups for all figures. mice. As shown in Fig. 3, treatment with niacin led to a
Values of P , 0.05 were considered significant. significant reduction in serum TC, LDL, and HDL, with
a characteristic of high ratio of HDL/LDL in mice fed an
HFD, but Tg remained unchanged in respect to vehicle-
RESULTS treated mice (Fig. 3A). The effect of niacin on TC, LDL,
HDL, and the ratio of HDL/LDL was not observed in
GPR109A-deficient mice are susceptible to DIO
Gpr109a2/2 mice (Fig. 3B). We also performed a GTT and
To explore GPR109A function in lipid metabolism, parallel ITT and documented that niacin-treated mice were less
experiments in mice with GPR109A (WT) and without glucose tolerant but exhibited insulin tolerance compara-
GPR109A (Gpr109a2/2) were performed. We recorded ble to control mice (Fig. 3C). No changes on GTT and ITT
weight gain weekly when both WT and Gpr109a2/2 male were detected in niacin-treated Gpr109a2/2 mice (Fig. 3D).
mice were fed either a NCD or HFD for 12 wk. As the Moreover, quantitative analysis showed that niacin treat-
feeding progressed, starting from ;10–12 wk of age, the ment results in a significant reduction in blood insulin (Fig.
GPR109A-knockout mice exhibited significantly increased 3E); however, HFD-fed mice treated with niacin exhibited
body weight compared with the WT mice, without any a marked decrease in HOMA-IR values compared to
significant difference in food intake between the two vehicle-treated mice (Fig. 3F). As expected, GPR109A-
groups (Fig. 1A– F). After 12 wk of NCD feeding, adipose knockout mice exhibited significantly higher fasting glu-
tissue weights and organ weights were examined. Our cose and insulin levels compared with the WT mice, which
data demonstrated that GPR109A-deficient mice dis- inevitably resulted in higher HOMA-IR levels (Supple-
played a significant increase in adipose tissue weights mental Fig. S2A–C), whereas no significant difference in
[including epididymal WAT (eWAT), perirenal WAT fasting glucose and insulin levels (including HOMA-IR
(pWAT), and inguinal WAT] and liver weight but not in values) was observed in niacin- and vehicle-treated
BAT or other organ weights (kidney, lung, heart, and Gpr109a2/2 mice when fed an HFD (Supplemental Fig.
spleen) when compared to WT mice (Fig. 1G, H and Sup- S2D–F). Taken together, these results suggest that niacin is
plemental Fig. S1A, B). Moreover, H&E-stained histologic likely to play a beneficial role in insulin sensitivity via
sections of eWAT and electron microscope observation of GPR109A.
liver and muscle revealed that GPR109A-deficient mice
had a striking visible increase in the amount and size of
lipid droplets in liver, muscle, and eWAT compared with GPR109A inhibits de novo lipogenesis in liver
the WT group (Fig. 1I). Taken together, it is more likely that in response to niacin
GPR109A plays an essential role in lipid metabolism.
Our data clearly showed that upon activation by niacin,
GPR109A exerts its inhibitory effects on lipid accumula-
GPR109A exerts inhibitory effects on HFD- tion in adipose tissue, liver, and muscle when fed an HFD.
induced obesity upon activation by niacin Moreover, quantitative analysis revealed that HFD-fed
mice displayed a significant increase in hepatic Tg content,
To further validate the role of GPR109A in the antiobesity whereas niacin supplement led to a marked reduction in
action, WT, and Gpr109a2/2 mice were fed a 6-wk HFD to Tg content in the liver; in contrast, no difference in TC and
induce obesity. A HFD supplemented with niacin (HFD + FFA levels was observed (Fig. 4A). In addition, the de-
niacin, 50 mM) or vehicle (HFD + water) was then in- crease in the hepatic insulin level was observed in mice
troduced during the following 8 wk. In WT mice groups, with niacin treatment (Fig. 4B), coincident with the result
treatment with niacin resulted in a significant reduction in serum (Fig. 3E). However, animals remain unaltered in
in body weight, but weekly food intake remained un- food intake. We then investigated whether or not niacin
changed as compared to vehicle-treated mice (Fig. 2A, B). plays a role in the regulation of hepatic de novo lipogenesis
As anticipated, the mice maintained on the HFD exhibited via GPR109A. We first used qRT-PCR to quantitatively
a progressive increase in body weight gain (Fig. 2A, C). analyze the GPR109A expression in mouse liver.
While lack of GPR109A prevented antiobesity actions of It has been reported that GPR109A is highly expressed
niacin (Fig. 2C, D), mice fed a HFD in response to treatment in adipocytes and activated immune cells (11, 14–16). Our

4768 Vol. 33 April 2019 The FASEB Journal x www.fasebj.org YE ET AL.


Figure 1. GPR109A-deficient animals display obesity. A) Body weights were recorded in mice fed a NCD (n = 7–8). B) Food intake
was recorded. C ) Representative images of fat mass are shown. D) Body weights were recorded in mice fed an HFD (n = 7–8). E )
Food intake was recorded. F ) Representative images of fat mass are shown. G) Fat pads and organ weights were recorded in mice
fed a NCD (n = 7–8). H ) Fat pads and organ weights were recorded in mice fed an HFD (n = 7–8). I ) Representative SEM images
in the liver (left panels; scale bars, 2 mm) and muscle (middle panels; scale bars, 0.5 mm) and H&E staining of eWAT (right
panels; scale bars, 100 mm) (n = 3 mice/group). iWAT, inguinal WAT; ns, not significant; WT, wild-type. All the data are
presented as means 6 SEM. Significance determined by unpaired, 2-tailed Student’s t test. *P , 0.05, **P , 0.01, ***P , 0.001,
****P , 0.0001.

GPR109A REGULATES ENERGY HOMEOSTASIS 4769


Figure 2. GPR109A exerts inhibitory effects on HFD-induced obesity upon activation by niacin. A–D) Body weight (A) and food
intake (B) were recorded weekly in WT mice (n = 10–11); body weight (C ) and food intake (D) were recorded weekly in
Gpr109a2/2 mice (n = 10). E ) Fat pads and organ weights were recorded after WT mice were anesthetized (n = 10–11). F ) Fat
pads and organ weights were recorded after Gpr109a2/2 mice were anesthetized (n = 10). G) Representative SEM images in the
liver (left panels; scale bars, 5 mm) and muscle (middle panels; scale bars, 0.5 mm) and H&E staining of eWAT (right panels; scale
bars, 100 mm) (n = 3 mice/group). iWAT, inguinal WAT; NA, niacin; ns, not significant; WT, wild type. All the data are presented as
means 6 SEM. Significance was determined by unpaired, 2-tailed Student’s t test. *P , 0.05, **P , 0.01, ****P , 0.0001.

data clearly showed that when fed an HFD, mice exhibited expression in liver or other tissues, except in BAT (Fig. 4D
a significant increase in lipid accumulation in the liver, and Supplemental Fig. S3A). In contrast, GPR109A was
whereas niacin supplementation resulted in a significant completely absent in the liver from Gpr109a2/2 mice
reduction of lipid accumulation in the liver. To confirm the (Supplemental Fig. S3B,C). GPR109A expression in the
role of GPR109A in the liver, we examined the expression liver was also confirmed at the protein level by Western
of GPR109A in different tissues and organs. High-level blot (Fig. 4E).
expression of GPR109A mRNA was detected in eWAT However, our data on GPR109A expression in mouse
and BAT (Fig. 4C). Interestingly, although relatively lower liver could not discriminate between the hepatocytes and
basal expression of GPR109A was detected, upon in- Kupffer cells. Therefore, to confirm the expression of
troduction of an HFD, GPR109A mRNA was markedly GPR109A in hepatocytes, we further analyzed the ex-
up-regulated in the liver but not in other organs (Fig. 4C, pression of GPR109A in the mice primary hepatocytes. As
D and Supplemental Fig. S3A). Moreover, niacin sup- anticipated, endogenous expression of GPR109A in both
plementation showed no significant effect on GPR109A mRNA and protein levels was detected in the primary

4770 Vol. 33 April 2019 The FASEB Journal x www.fasebj.org YE ET AL.


Figure 3. Niacin improves the dyslipoidemia and insulin sensitivity via GPR109A. A) Serum lipid parameters, including total Tg,
TC, HDL, and LDL were measured in WT mice using biochemical testing kits (n = 7–8). B) Serum lipid parameters were
measured in Gpr109a2/2 mice (n = 7–8). C ) GTT and ITT results and the corresponding AUC in WT mice (n = 5–6). D) GTT
and ITT results and the corresponding AUC in Gpr109a2/2 mice (n = 5–6). E ) Serum insulin level during GTT was measured by
ELISA in WT mice (n = 5–6). F ) HOMA-IR index was calculated in WT mice as stated in the Materials and Methods section (n =
5–6). All the data are presented as mean 6 SEM. NA, niacin; ns, not significant; WT, wild type. Significance was determined by
unpaired, 2-tailed Student’s t test. *P , 0.05, **P , 0.01, ***P , 0.0001.

hepatocytes (Fig. 4F). To confirm the role of GPR109A in the enzyme activities of ACC, ACC1, and PDH in the liver,
the regulation of de novo lipogenesis in the liver, we used whereas no change in eWAT was observed (Fig. 4G).
ELISA to quantitatively analyze the activities of key en- However, in Gpr109a2/2 mice, no difference in the enzyme
zymes involved in fatty acid biosynthesis, ACC, ACC1, activities of PDH and ACC in the liver following treat-
and PDH (32). As shown in Fig. 4G, in WT mice fed a HFD, ment with niacin was detected (Fig. 4H). Our data also
treatment with niacin resulted in a significant decrease in showed that niacin supplementation led to no statistically

GPR109A REGULATES ENERGY HOMEOSTASIS 4771


Figure 4. Activated GPR109A directly inhibits hepatocyte lipogenesis. A) Liver lipid content, including the levels of Tg, TC, and
FFA, were measured (n = 5–6). B) ELISA analysis revealed the insulin level reduced significantly in the mice treated with niacin
(n = 5). C ) GPR109A gene expression was measured by qRT-PCR in muscle, liver, intestine, eWAT, and BAT of the mice fed a
NCD, HFD, or HFD with 50 mM niacin, respectively (n = 8–10). Gene expression in muscle served as the statistical controls. D)
Expression of GPR109A was measured in the liver isolated from the WT mice fed a NCD or HFD with or without niacin by
(continued on next page)

4772 Vol. 33 April 2019 The FASEB Journal x www.fasebj.org YE ET AL.


significant changes in the activity of DGAT2 (Fig. 4G), improved cristae organization in HFD-fed WT mice (Fig.
which directly catalyzes the formation of Tgs from diac- 5D). Furthermore, we assayed the expression of genes
ylglycerol and Acyl-CoA in fat, liver, and skin cells (33). In involved in BAT thermogenesis by real time PCR analysis.
addition, these data were further confirmed by quan- As shown in Fig. 5E, the mRNA expression of uncoupling
titative determination of mRNA transcripts of sterol reg- protein 1 (UCP1), a critical player in allowing electrons to
ulatory element–binding protein-1c (SREBP-1c), a key be released as heat rather than stored, and PR domain
transcriptional activator of lipogenic genes and its down- containing 16 (PRDM16), a well-known BAT tran-
stream target genes, ACC1, fatty acid synthase (FAS), and scriptional regulator, were significantly up-regulated in
stearoyl-CoA desaturase-1 (SCD-1) by real time RT-PCR. HFD-fed WT mice in response to treatment with niacin.
As revealed in Fig. 4I, administration of niacin resulted in a However, no difference was detected in niacin-treated
significant decrease in mRNA levels of SREBP-1c and its Gpr109a2/2 mice (Fig. 5F). Additionally, we also ob-
target genes, ACC1, FAS, and SCD-1 in the liver compared served that niacin showed the potential to elevate the
to the vehicle-treated mice. Our data suggest the inhibitory expression of beige-specific genes including Cd137, Tbx1,
effects of niacin-GPR109A on hepatic de novo lipogenesis; and Tmem26 in subcutaneous WAT (sWAT) and the ex-
however, detailed mechanism involved in this inhibition pression of peroxisome proliferator-activated receptor g
needs further exploration. coactivator 1-a (PGC-1a) and Tbx1 in eWAT but not in
pWAT in HFD-fed WT mice (Fig. 5G) or Gpr109a2/2 mice
(Fig. 5H). These data collectively suggest that nia-
Agonist-activated GPR109A triggers BAT cin stimulates energy expenditure through GPR109A in
and/or browning WAT thermogenic activity HFD-fed mice.

Obesity is an abnormal accumulation of lipid in adipose


tissue caused by an imbalance between energy intake and Niacin-activated GPR109A leads to a reduction
expenditure. In the HFD-induced obesity mouse model, in dietary fatty acid absorption
niacin supplementation led to a significant reduction in
body weight but no change in daily food intake, indicating Recent studies have demonstrated that GPR109A is
that niacin is more likely to promote energy expenditure. expressed in colonic and intestinal epithelial cells and
To validate this hypothesis, we first monitored the core mediates the local control of intestinal glucose absorp-
body temperature by measuring the anal temperature of tion (34) and the tumor-suppressive effects of the bac-
mice fed an HFD and treated with vehicle or niacin upon terial fermentation product butyrate in the colon (35).
acute cold exposure. GPR109A-deficient mice showed a Accordingly, we further examined the role of GPR109A
continuous decline in body temperature and reached 32°C expression in colonic and intestinal epithelial cells in the
after acute cold exposure at 4°C for 6 h, whereas WT mice control of intestinal fatty acid absorption. We collected
were able to maintain their body temperature at 33.5°C feces from mice housed individually from 5 consecutive
(Fig. 5A). Moreover, WT mice fed an HFD and treated days and measured fecal lipid contents. As indicated in
with niacin revealed significantly higher body tem- Fig. 6, the weights of feces normalized to body weight in
peratures than that of vehicle-treated mice; however, no the niacin-treated mice when fed a HFD compared to
significant difference was observed between the niacin- the control mice (Fig. 6A). Further analysis showed that
and vehicle-treated group in GPR109A-deficient mice total lipid, Tg, TC, and FFA in feces were significantly
in response to acute cold exposure (Fig. 5A) and in WT increased when HFD-fed mice treated with niacin were
mice without acute cold exposure (Fig. 5B). These data compared to the control mice (Fig. 6A). However, total
collectively confirm cold tolerance in niacin-treated WT lipid in feces was reduced in HFD-fed Gpr109a2/2 mice
mice. treated with niacin; no difference in Tg, TC, and FFA
BAT is the major site essential to generate heat (ther- was observed between niacin-treated mice and control
mogenesis) for the maintenance of body temperature. mice (Fig. 6B). This was confirmed by RT-PCR quanti-
Therefore, we examined the ultrastructure of BAT tative analysis of 3 key proteins [fatty acid transport
mitochondria. Transmission electron microscopy dem- protein 4 (FATP4), intestinal-type fatty acid–binding
onstrated that BAT from Gpr109a2/2 mice exhibited a protein (I-FABP), and NCP1L1] involved in the control
significant reduction in the number and size of mito- of fatty acid and cholesterol absorption. The mRNA
chondria with aberrant cristae compared to WT mice (Fig. levels of the 3 genes were obviously decreased in the
5C). In addition, niacin treatment caused a remark- WT niacin group but not in the Gpr109a2/2 mice (Fig.
able increase in number and size of mitochondria with 6C). These findings suggest that niacin is likely to

quantitative analyses (n = 8–10). E ) The relative gene expression of GPR109A in the liver was calculated by Western blot (n = 4).
F ) Expression of GPR109A was measured in primary mice hepatocytes by semiquantification PCR (left panel) and Western blot
(right panel) (n = 3). G) Comparing the activities of enzymes involving in lipogenesis (PDH, ACC, ACC1, and DGAT2) of WT
mice fed an HFD with or without niacin in the liver and eWAT (n = 5). H ) The activities of PDH and ACC in the liver were
measured in Gpr109a2/2 mice fed an HFD (n = 7, 9). I ) The mRNA expression analysis of genes involved in de novo lipogenesis in
the liver, and the data of the WT (vehicle) group served as the statistical controls (n = 8). NA, niacin; ns, not significant; WT, wild-
type. All data are expressed as means 6 SEM. ANOVA followed by unpaired, 2-tailed Student’s t test. Significant differences
between groups are indicated with asterisks. *P , 0.05, **P , 0.01, ***P , 0.001.

GPR109A REGULATES ENERGY HOMEOSTASIS 4773


Figure 5. Niacin treatment triggers BAT thermogenesis via GPR109A. A) To test resistance to cold exposure, mice were exposed
to 4°C for 6 h; anal temperature monitored hourly (n = 5, 6). B) Anal temperature monitored weekly under room temperature
(n = 5). C ) Electron micrographs of the BAT in WT and Gpr109a2/2 mice. Scale bars, 1 mm. D) Electron micrographs of the
BAT at the end of niacin treatment. Scale bars, 1 mm. E ) The mRNA expression of UCP1, PGC-1a, and PRDM16 in BAT of WT
mice (n = 10, 11). F ) The mRNA expression of UCP1, PGC-1a, and PRDM16 in Gpr109a2/2 mice (n = 6, 7). G) The mRNA
expression of brown or beige genes (UCP1, PRDM16, PGC-1a, Cd137, Tbx1, and Tmem26) in pWAT, sWAT and eWAT in WT
mice (n = 10, 11). H ) The mRNA expression of brown or beige genes in pWAT, sWAT, and eWAT in Gpr109a2/2 mice (n = 6,
7). NA, niacin; ns, not significant; WT, wild type. All values are presented as mean 6 SEM. ANOVA followed by unpaired, 2-
tailed Student’s t test. Significant differences between groups are indicated with asterisks. *P , 0.05, **P , 0.01, ***P , 0.001.

4774 Vol. 33 April 2019 The FASEB Journal x www.fasebj.org YE ET AL.


Figure 6. Activated GPR109A decreases fatty acid absorption. Feces were collected over a period of 5 d from WT mice (n =
7, 9) as well as Gpr109a 2/2 mice (n = 6) fed an HFD. A) Fecal mass was normalized to body weight (mg/g). Fecal lipids
were extracted, and the fecal lipid concentration was expressed as milligrams of lipids per gram of feces weight. Total Tg,
TC, and FFA were measured in WT mice. B) Feces lipid indicators were measured in Gpr109a 2/2 mice. C ) We performed
qRT-PCR using total mRNA isolated from intestines and examined the mRNA expression of indicated genes involved
in fatty acid absorption (FATP4 and I-FABP) and cholesterol absorption (NPC1L1). The data from the vehicle group
served as the statistical controls. NA, niacin; ns, not significant; WT, wild type. All values are presented as mean 6 SEM.
ANOVA followed by 2-tailed Student’s t test. Significant differences between groups are indicated with asterisks. *P , 0.05,
**P , 0.01.

exhibit the potential to inhibit the dietary fat absorption energy expenditure, and intestinal fatty acid absorption
through GPR109A, thereby causing the decrease in fat through GPR109A.
deposit and body weight. During the breeding process of GPR109A-knockout
mice obtained from Dr. Offermanns, we observed that
the deletion of GPR109A resulted in obvious increases in
DISCUSSION body weight and fat mass compared to WT mice. In
contrast, lactate receptor GPR81-deficient mice gain
Since the identification of GPR109A, a Gi/o-coupled significantly less body weight than WT mice under HFD-
receptor, as the molecular target for niacin (20, 22, 36), feeding conditions, although Gi/o-coupled GPR81 ex-
most studies have focused on its pronounced hypo- erts the same activity in the inhibition of lipolysis in
lipidemic and cardioprotective action. Recent investi- WAT (41). This prompted us to initiate this study to
gations have also shed light on the anti-inflammatory evaluate the exact role of GPR109A in the regulation of
and anti-oxidative effects of GPR109A (22, 37–40). energy homeostasis. Recent studies indicated that niacin
However, little attention has been paid to the potential treatment remarkably reduced body weight in the obese
of this receptor to favorably modulate de novo lipogen- mice (42). Previous investigations have demonstrated
esis and energy homeostasis. In the present study, we that the ketone body b-hydroxybutyrate (bOHB), as an
explored the physiologic roles of niacin-GPR109A in important metabolic intermediate and also a gut bacte-
the regulation of lipid metabolism and energy homeo- rial fermentation-yielded product, which is the only
stasis in an HFD-induced murine model of obesity known endogenous ligand for GPR109A (18), causes
combined with GPR109A-deficient mice. The most reductions of body weight and fat content (43, 44).
important observation is that niacin supplementation Moreover, niacin and bOHB were also found to improve
prevented the progression of dietary obesity in HFD- the alcoholic or nonalcoholic fatty liver disease in rats
fed WT mice but not in HFD-fed GPR109A-deficient and mice (45–51). Using a combination of GPR109A-
mice. Mechanistically, we demonstrated that niacin- deficient mice with HFD-induced model of obesity, we
mediated reduction in body weight and fat mass was provided further evidence that niacin treatment led to a
caused by fine-tuning of de novo lipogenesis in liver, significant reduction in body weight and fat mass

GPR109A REGULATES ENERGY HOMEOSTASIS 4775


through GPR109A when fed a NCD or HFD. These Interestingly, we found that mitochondria in BAT of
findings suggest that GPR109A has the potential to GPR109A knockout mice were smaller in size with ab-
control energy homeostasis, preventing weight gain. normal cristae structures compared to WT mice, and
Our results clearly revealed that niacin treatment re- niacin treatment led to significant improvement in mi-
markably reduced liver weight in WT mice fed an HFD but tochondrial size and cristae structure of mice fed an
not in GPR109A-deficient mice. Ultrastructural examina- HFD. Additionally, niacin stimulated a beige fat ther-
tion validated that GPR109A-deficient mice had a striking mogenic program of the sWAT and eWAT. These data
visible increase in the amount and size of lipid droplets in strongly suggest that niacin treatment triggers BAT
the liver compared with the WT group, and niacin treat- and/or browning WAT thermogenic activity in mice
ment led to a significant reduction in the amount and size through GPR109A.
of lipid droplets in the liver of WT mice when fed an HFD. Importantly, GPR109A has been shown to functionally
These findings suggest that GPR109A plays an important express in colonic and intestinal epithelial cells (34, 35).
role in the regulation of de novo lipogenesis in liver. The Niacin has been found to locally control glucose uptake at
liver has long been recognized as the major site of de novo the level of the small intestine through GPR109A (34). This
lipogenesis, responsible for the conversion of excess di- prompts us to examine whether or not GPR109A is in-
etary carbohydrate into Tgs. De novo lipogenesis turnover volved in the control of uptake of sterols and fatty acids in
accounts for ;20% of lipid turnover within adipose tissue the level of the small intestine. To our surprise, niacin
(52); however, de novo lipogenesis in nonadipose tissues supplementation caused a significant increase of total
leads to ectopic lipid accumulation responsible for meta- lipids, Tgs, cholesterols, and FFAs in the feces of WT mice
bolic stress and insulin resistance (53). Although expres- fed an HFD, but no change was observed in HFD-fed
sion of GPR109A in the liver remains controversial, the GPR109A-deficient mice. Further gene expression anal-
abundance of GPR109A mRNA detected was higher in the ysis showed that genes encoding FATP4, I-FABP, and
liver in lean-type than in fat-type cows (54). GPR109A has Niemann-Pick C1-Like 1 (NPC1L1) exhibited a significant
been found to be expressed in the murine liver, but the reduction in expression in the intestinal tissues. It is estab-
basal expression is low (55). In the current study, we pro- lished that polytopic transmembrane protein NPC1L1 is
vided evidence to confirm that GPR109A mRNA and essential for intestinal absorption of cholesterol (66),
protein were detected in the murine liver at a relatively whereas proteins FATP4 and I-FABP play a role in dietary
lower level; however, mice fed an HFD exhibited signifi- fat absorption and fatty acid transport, respectively (67,
cantly enhanced expression of GPR109A in the liver. In 68). Taken together, these findings suggest that in addi-
addition, the expression of GPR109A in primary mice tion to the control of glucose uptake, GPR109A expressed
hepatocytes was also detected. Further biochemical anal- in the small intestine is likely involved in the local regu-
ysis of key enzymes involved in lipogenesis demonstrated lation of intestinal absorption of Tgs, cholesterols, and
that niacin exhibited inhibitory effects on PDH (the gate- FFAs.
keeping enzyme of pyruvate flux into acetyl-CoA) and Consistent with previous studies (43, 69–71), we also
ACC1 (the key regulatory step of fatty acid synthesis) (56, observed that the HFD-induced obese mice treated with
57). These findings were verified by real-time qRT-PCR for niacin exhibited an obvious reduction in serum insu-
genes encoding SREBP-1c (a key transcriptional activator lin concentration and glucose tolerance compared to
of lipogenic genes) and lipogenic enzymes (ACC1, FAS, the vehicle-treated mice. However, our data demon-
and SCD-1) (58–60). Niacin has been shown to directly strated that treatment with niacin led to a significant im-
inhibit the activity of DGAT2, the key enzyme that cata- provement in insulin resistance in HFD-fed obese mice
lyzes the final step in Tg synthesis in the liver (30, 61). (HOMA-IR). GPR109A has been shown to be functionally
However, our results strongly suggest that niacin ex- expressed in both human and murine islet b-cells (72, 73).
erts its inhibitory effects on hepatic lipogenesis through Chen et al. (42) have shown that niacin exerts its inhibitory
GPR109A. actions on b-cell function via GPR109A-mediated signal-
It is noteworthy that mice treated with niacin ing but without affecting islet architecture. Nevertheless, a
exhibited a remarkable reduction in body weight and fat recent study demonstrated that GPR109A expression was
mass without affecting food intake, consistent with down-regulated dramatically in islet b-cells of type 2 di-
previous observations that bOHB supplementation eli- abetic patients as well as in diabetes/diabetes mice, sug-
cited body weight loss but without a decrease in food gesting that GPR109A signaling plays an important role in
intake (43, 44). It suggests that energy expenditure is the prevention of diabetes (72). Moreover, an early study
likely to contribute to niacin-induced body weight loss. observed that a short-term niacin analog administration
Results derived from cold response experiments showed resulted in an improved glucose tolerance in prediabetic or
that thermogenic function was enhanced in WT mice diabetic patients (74). In addition, our data clearly dem-
treated with niacin but not in GPR109A knockout mice. onstrated that niacin treatment caused a significant re-
Real-time PCR analysis demonstrated that upon treat- duction in hepatic de novo lipogenesis and ectopic lipid
ment with niacin, the expression of thermogenic genes accumulation in the liver, adipose tissue, and muscle of
UCP1 (a thermogenic protein exclusively expressed mice fed an HFD, leading to a beneficial effect on insulin
in brown or beige adipocytes) (62, 63), PGC-1a (a resistance. The effects of niacin on glucose homeostasis
master regulator of mitochondrial biogenesis) (64), and remain controversial, and the exact roles of GPR109A in
PRDM16 (a well-known BAT transcriptional regulator) the regulation of insulin resistance now deserve thorough
(65) were significantly increased in mice fed an HFD. evaluation.

4776 Vol. 33 April 2019 The FASEB Journal x www.fasebj.org YE ET AL.


In summary, the data presented in this report have Sturua, L., Sykes, B. L., Tobias, M., Tran, B. X., Trasande, L.,
demonstrated the novel role of niacin in the regulation of Toyoshima, H., van de Vijver, S., Vasankari, T. J., Veerman, J. L.,
Velasquez-Melendez, G., Vlassov, V. V., Vollset, S. E., Vos, T., Wang,
energy homeostasis by fine-tuning of hepatic de novo C., Wang, X., Weiderpass, E., Werdecker, A., Wright, J. L., Yang,
lipogenesis, energy expenditure, and intestinal fat ab- Y. C., Yatsuya, H., Yoon, J., Yoon, S. J., Zhao, Y., Zhou, M., Zhu, S.,
sorption via GPR109A-mediated signaling. Our findings Lopez, A. D., Murray, C. J., and Gakidou, E. (2014) Global, regional,
and national prevalence of overweight and obesity in children and
provide a critical clue to the potential new therapeutic
adults during 1980-2013: a systematic analysis for the Global Burden
strategies that could treat obesity, diabetes, and car- of Disease Study 2013. Lancet 384, 766–781
diovascular disease. As the debate continues about 2. Wang, Y. C., McPherson, K., Marsh, T., Gortmaker, S. L., and Brown,
the favorable effects of niacin-GPR109A on the devel- M. (2011) Health and economic burden of the projected obesity
trends in the USA and the UK. Lancet 378, 815–825
opment and progression of atherosclerosis and its 3. Olsen, M. K., Johannessen, H., Cassie, N., Barrett, P., Takeuchi, K.,
complications, it will be vital to further perform a more Kulseng, B., Chen, D., and Zhao, C. M. (2017) Steady-state energy
comprehensive assessment of niacin-GPR109A in the balance in animal models of obesity and weight loss. Scand. J. Gastro-
regulation of energy homeostasis in addition to anti- enterol. 52, 442–449
4. Lowell, B. B., and Spiegelman, B. M. (2000) Towards a molecular
inflammation. understanding of adaptive thermogenesis. Nature 404, 652–660
5. Haslam, D. W., and James, W. P. (2005) Obesity. Lancet 366,
1197–1209
ACKNOWLEDGMENTS 6. Kopelman, P. G. (2000) Obesity as a medical problem. Nature 404,
635–643
7. Cunningham, J. W., and Wiviott, S. D. (2014) Modern obesity
The authors thank Dr. Stefan Offermanns (Max Planck pharmacotherapy: weighing cardiovascular risk and benefit. Clin.
Institute for Heart and Lung Research, Bad Nauheim, Germany) Cardiol. 37, 693–699
for providing GPR109A KO (C57BL) breeders and critical 8. Moreira, F. A., Grieb, M., and Lutz, B. (2009) Central side-effects of
comments. Junying Li (Center of Analysis and Measurement, therapies based on CB1 cannabinoid receptor agonists and antago-
Zhejiang University), Qiong Huang (Core Facilities, School of nists: focus on anxiety and depression. Best Pract. Res. Clin. Endocrinol.
Medicine, Zhejiang University), and Shenghui Hong (Laboratory Metab. 23, 133–144
Animal Center, Zhejiang University) are acknowledged for their 9. Manning, S., Pucci, A., and Finer, N. (2014) Pharmacotherapy for
technical assistance. This work was supported by grants from the obesity: novel agents and paradigms. Ther. Adv. Chronic Dis. 5, 135–148
10. Altschul, R., Hoffer, A., and Stephen, J. D. (1955) Influence of
Ministry of Science and Technology of China (2012CB910402 to
nicotinic acid on serum cholesterol in man. Arch. Biochem. Biophys. 54,
N.Z.), the National Natural Science Foundation of China (81173106, 558–559
31200621, and 31771543 to N.Z. and Y.S.), the Zhejiang Provincial 11. Carlson, L. A. (2005) Nicotinic acid: the broad-spectrum lipid drug. A
Natural Science Foundation of China (LY15C050001 to N.Z.), and 50th anniversary review. J. Intern. Med. 258, 94–114
the Siyuan Foundation (to N.Z. and Y.S.). The authors declare no 12. Guyton, J. R., Goldberg, A. C., Kreisberg, R. A., Sprecher, D. L.,
conflicts of interest. Superko, H. R., and O’Connor, C. M. (1998) Effectiveness of once-
nightly dosing of extended-release niacin alone and in combination
for hypercholesterolemia. Am. J. Cardiol. 82, 737–743
13. Elam, M. B., Hunninghake, D. B., Davis, K. B., Garg, R., Johnson, C.,
AUTHOR CONTRIBUTIONS Egan, D., Kostis, J. B., Sheps, D. S., and Brinton, E. A. (2000) Effect of
niacin on lipid and lipoprotein levels and glycemic control in patients
Y. Shi and N. Zhou designed and supervised the research; with diabetes and peripheral arterial disease: the ADMIT study: a
L. Ye, Z. Cao, X. Lai, and W. Wang performed in vivo randomized trial. Arterial Disease Multiple Intervention Trial. JAMA
284, 1263–1270
experiments; L. Ye, Z. Cao, X. Lai, W. Wang, Z. Guo, 14. Soga, T., Kamohara, M., Takasaki, J., Matsumoto, S., Saito, T., Ohishi,
L. Yan, and Y. Wang performed in vitro experiments; and T., Hiyama, H., Matsuo, A., Matsushime, H., and Furuichi, K. (2003)
L. Ye, Y. Shi, and N. Zhou contributed to data interpretation Molecular identification of nicotinic acid receptor. Biochem. Biophys.
and wrote the manuscript. Res. Commun. 303, 364–369
15. Tunaru, S., Kero, J., Schaub, A., Wufka, C., Blaukat, A., Pfeffer, K., and
Offermanns, S. (2003) PUMA-G and HM74 are receptors for nic-
otinic acid and mediate its anti-lipolytic effect. Nat. Med. 9,
REFERENCES 352–355
16. Wise, A., Foord, S. M., Fraser, N. J., Barnes, A. A., Elshourbagy, N.,
1. Ng, M., Fleming, T., Robinson, M., Thomson, B., Graetz, N., Eilert, M., Ignar, D. M., Murdock, P. R., Steplewski, K., Green, A.,
Margono, C., Mullany, E. C., Biryukov, S., Abbafati, C., Abera, S. F., Brown, A. J., Dowell, S. J., Szekeres, P. G., Hassall, D. G., Marshall,
Abraham, J. P., Abu-Rmeileh, N. M., Achoki, T., AlBuhairan, F. S., F. H., Wilson, S., and Pike, N. B. (2003) Molecular identification of
Alemu, Z. A., Alfonso, R., Ali, M. K., Ali, R., Guzman, N. A., Ammar, high and low affinity receptors for nicotinic acid. J. Biol. Chem. 278,
W., Anwari, P., Banerjee, A., Barquera, S., Basu, S., Bennett, D. A., 9869–9874
Bhutta, Z., Blore, J., Cabral, N., Nonato, I. C., Chang, J. C., 17. Offermanns, S., Colletti, S. L., Lovenberg, T. W., Semple, G., Wise, A.,
Chowdhury, R., Courville, K. J., Criqui, M. H., Cundiff, D. K., and IJzerman, A. P. (2011) International union of basic and clinical
Dabhadkar, K. C., Dandona, L., Davis, A., Dayama, A., Dharmaratne, pharmacology. LXXXII: nomenclature and classification of hydroxy-
S. D., Ding, E. L., Durrani, A. M., Esteghamati, A., Farzadfar, F., Fay, carboxylic acid receptors (GPR81, GPR109A, and GPR109B). Phar-
D. F., Feigin, V. L., Flaxman, A., Forouzanfar, M. H., Goto, A., Green, macol. Rev. 63, 269–290
M. A., Gupta, R., Hafezi-Nejad, N., Hankey, G. J., Harewood, H. C., 18. Taggart, A. K., Kero, J., Gan, X., Cai, T. Q., Cheng, K., Ippolito, M.,
Havmoeller, R., Hay, S., Hernandez, L., Husseini, A., Idrisov, B. T., Ren, N., Kaplan, R., Wu, K., Wu, T. J., Jin, L., Liaw, C., Chen, R.,
Ikeda, N., Islami, F., Jahangir, E., Jassal, S. K., Jee, S. H., Jeffreys, M., Richman, J., Connolly, D., Offermanns, S., Wright, S. D., and Waters,
Jonas, J. B., Kabagambe, E. K., Khalifa, S. E., Kengne, A. P., Khader, M. G. (2005) (D)-beta-hydroxybutyrate inhibits adipocyte lipolysis via
Y. S., Khang, Y. H., Kim, D., Kimokoti, R. W., Kinge, J. M., Kokubo, Y., the nicotinic acid receptor PUMA-G. J. Biol. Chem. 280, 26649–26652
Kosen, S., Kwan, G., Lai, T., Leinsalu, M., Li, Y., Liang, X., Liu, S., 19. Digby, J. E., Lee, J. M., and Choudhury, R. P. (2009) Nicotinic acid and
Logroscino, G., Lotufo, P. A., Lu, Y., Ma, J., Mainoo, N. K., Mensah, G. A., the prevention of coronary artery disease. Curr. Opin. Lipidol. 20,
Merriman, T. R., Mokdad, A. H., Moschandreas, J., Naghavi, M., 321–326
Naheed, A., Nand, D., Narayan, K. M., Nelson, E. L., Neuhouser, 20. Lukasova, M., Malaval, C., Gille, A., Kero, J., and Offermanns, S.
M. L., Nisar, M. I., Ohkubo, T., Oti, S. O., Pedroza, A., Prabhakaran, (2011) Nicotinic acid inhibits progression of atherosclerosis in mice
D., Roy, N., Sampson, U., Seo, H., Sepanlou, S. G., Shibuya, K., Shiri, through its receptor GPR109A expressed by immune cells. J. Clin.
R., Shiue, I., Singh, G. M., Singh, J. A., Skirbekk, V., Stapelberg, N. J., Invest. 121, 1163–1173

GPR109A REGULATES ENERGY HOMEOSTASIS 4777


21. Wu, B. J., Yan, L., Charlton, F., Witting, P., Barter, P. J., and Rye, K. A. butyrate, suppresses colonic inflammation and carcinogenesis.
(2010) Evidence that niacin inhibits acute vascular inflammation and Immunity 40, 128–139
improves endothelial dysfunction independent of changes in plasma 40. Bühler, S., Frahm, J., Tienken, R., Kersten, S., Meyer, U., Huber, K.,
lipids. Arterioscler. Thromb. Vasc. Biol. 30, 968–975 and Dänicke, S. (2018) Effects of energy supply and nicotinic acid
22. Digby, J. E., Martinez, F., Jefferson, A., Ruparelia, N., Chai, J., Wamil, supplementation on serum anti-oxidative capacity and on expression
M., Greaves, D. R., and Choudhury, R. P. (2012) Anti-inflammatory of oxidative stress-related genes in blood leucocytes of periparturient
effects of nicotinic acid in human monocytes are mediated by primi- and pluriparous dairy cows. J. Anim. Physiol. Anim. Nutr. (Berl.)
GPR109A dependent mechanisms. Arterioscler. Thromb. Vasc. Biol. 32, 102, e87–e98
669–676 41. Ahmed, K., Tunaru, S., Tang, C., Müller, M., Gille, A., Sassmann, A.,
23. Mitrofanov, V. A., Ovchinnikova, N. M., Belova, S. V., Fedotova, M. V., Hanson, J., and Offermanns, S. (2010) An autocrine lactate loop
and Gladkova, E. V. (2005) Inflammatory degeneration of joint tissue mediates insulin-dependent inhibition of lipolysis through GPR81.
in adjuvant arthritis after intraarticular treatment with the mixture of Cell Metab. 11, 311–319
silver drug and nicotinic acid. Bull. Exp. Biol. Med. 140, 702–704 42. Chen, L., So, W. Y., Li, S. Y., Cheng, Q., Boucher, B. J., and
24. Shi, Y., Lai, X., Ye, L., Chen, K., Cao, Z., Gong, W., Jin, L., Wang, C., Leung, P. S. (2015) Niacin-induced hyperglycemia is partially
Liu, M., Liao, Y., Wang, J. M., and Zhou, N. (2017) Activated niacin mediated via niacin receptor GPR109a in pancreatic islets. Mol.
receptor HCA2 inhibits chemoattractant-mediated macrophage mi- Cell. Endocrinol. 404, 56–66
gration via Gbg/PKC/ERK1/2 pathway and heterologous receptor 43. Davis, J. D., Wirtshafter, D., Asin, K. E., and Brief, D. (1981) Sustained
desensitization. Sci. Rep. 7, 42279 intracerebroventricular infusion of brain fuels reduces body weight
25. Cho, K. H., Kim, H. J., Rodriguez-Iturbe, B., and Vaziri, N. D. (2009) and food intake in rats. Science 212, 81–83
Niacin ameliorates oxidative stress, inflammation, proteinuria, and 44. Gao, Z., Yin, J., Zhang, J., Ward, R. E., Martin, R. J., Lefevre, M., Cefalu,
hypertension in rats with chronic renal failure. Am. J. Physiol. Renal W. T., and Ye, J. (2009) Butyrate improves insulin sensitivity and
Physiol. 297, F106–F113 increases energy expenditure in mice. Diabetes 58, 1509–1517
26. Shehadah, A., Chen, J., Zacharek, A., Cui, Y., Ion, M., Roberts, C., 45. Kolovou, G. D., Salpea, K. D., Mihas, C., Malakos, I., Kafaltis, N.,
Kapke, A., and Chopp, M. (2010) Niaspan treatment induces Bilianou, H. G., Adamopoulou, E. N., Mykoniatis, M., and Cokkinos,
neuroprotection after stroke. Neurobiol. Dis. 40, 277–283 D. V. (2008) Comparison of simvastatin and nicotinic acid
27. Kwon, W. Y., Suh, G. J., Kim, K. S., and Kwak, Y. H. (2011) Niacin administration in alcohol-treated Wistar rats. Hellenic J. Cardiol. 49,
attenuates lung inflammation and improves survival during sepsis by 79–85
downregulating the nuclear factor-kB pathway. Crit. Care Med. 39, 46. Hu, M., Chu, W. C., Yamashita, S., Yeung, D. K., Shi, L., Wang, D.,
328–334 Masuda, D., Yang, Y., and Tomlinson, B. (2012) Liver fat reduction
28. Rahman, M., Muhammad, S., Khan, M. A., Chen, H., Ridder, D. A., with niacin is influenced by DGAT-2 polymorphisms in hyper-
Müller-Fielitz, H., Pokorná, B., Vollbrandt, T., Stölting, I., Nadrowitz, triglyceridemic patients. J. Lipid Res. 53, 802–809
R., Okun, J. G., Offermanns, S., and Schwaninger, M. (2014) The 47. Dou, X., Shen, C., Wang, Z., Li, S., Zhang, X., and Song, Z. (2013)
b-hydroxybutyrate receptor HCA2 activates a neuroprotective subset Protection of nicotinic acid against oxidative stress-induced cell death
of macrophages. Nat. Commun. 5, 3944 in hepatocytes contributes to its beneficial effect on alcohol-induced
29. Chen, H., Assmann, J. C., Krenz, A., Rahman, M., Grimm, M., liver injury in mice. J. Nutr. Biochem. 24, 1520–1528
Karsten, C. M., Köhl, J., Offermanns, S., Wettschureck, N., and 48. Ganji, S. H., Kukes, G. D., Lambrecht, N., Kashyap, M. L., and
Schwaninger, M. (2014) Hydroxycarboxylic acid receptor 2 Kamanna, V. S. (2014) Therapeutic role of niacin in the prevention
mediates dimethyl fumarate’s protective effect in EAE. J. Clin. and regression of hepatic steatosis in rat model of nonalcoholic fatty
Invest. 124, 2188–2192 liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 306, G320–G327
30. Ganji, S. H., Tavintharan, S., Zhu, D., Xing, Y., Kamanna, V. S., and 49. Li, Q., Xie, G., Zhang, W., Zhong, W., Sun, X., Tan, X., Sun, X., Jia, W.,
Kashyap, M. L. (2004) Niacin noncompetitively inhibits DGAT2 but and Zhou, Z. (2014) Dietary nicotinic acid supplementation
not DGAT1 activity in HepG2 cells. J. Lipid Res. 45, 1835–1845 ameliorates chronic alcohol-induced fatty liver in rats. Alcohol. Clin.
31. Folch, J., Lees, M., and Sloane Stanley, G. H. (1957) A simple method Exp. Res. 38, 1982–1992
for the isolation and purification of total lipides from animal tissues. 50. Ganji, S. H., Kashyap, M. L., and Kamanna, V. S. (2015) Niacin inhibits
J. Biol. Chem. 226, 497–509 fat accumulation, oxidative stress, and inflammatory cytokine IL-8 in
32. Tong, L. (2005) Acetyl-coenzyme A carboxylase: crucial metabolic cultured hepatocytes: impact on non-alcoholic fatty liver disease.
enzyme and attractive target for drug discovery. Cell. Mol. Life Sci. 62, Metabolism 64, 982–990
1784–1803 51. Chen, Y., Ouyang, X., Hoque, R., Garcia-Martinez, I., Yousaf, M. N.,
33. Oelkers, P., Behari, A., Cromley, D., Billheimer, J. T., and Sturley, S. L. Tonack, S., Offermanns, S., Dubuquoy, L., Louvet, A., Mathurin, P.,
(1998) Characterization of two human genes encoding acyl coenzyme Massey, V., Schnabl, B., Bataller, R. A., and Mehal, W. Z. (2018)
A:cholesterol acyltransferase-related enzymes. J. Biol. Chem. 273, b-hydroxybutyrate protects from alcohol-induced liver injury via a
26765–26771 Hcar2-cAMP dependent pathway. J. Hepatol. 69, 687–696
34. Wong, T. P., Chan, L. K. Y., and Leung, P. S. (2015) Involvement of the 52. Strawford, A., Antelo, F., Christiansen, M., and Hellerstein, M. K.
niacin receptor GPR109a in the localControl of glucose uptake in (2004) Adipose tissue triglyceride turnover, de novo lipogenesis, and
small intestine of type 2Diabetic mice. Nutrients 7, 7543–7561 cell proliferation in humans measured with 2H2O. Am. J. Physiol.
35. Thangaraju, M., Cresci, G. A., Liu, K., Ananth, S., Gnanaprakasam, Endocrinol. Metab. 286, E577–E588
J. P., Browning, D. D., Mellinger, J. D., Smith, S. B., Digby, G. J., 53. Solinas, G., Borén, J., and Dulloo, A. G. (2015) De novo lipogenesis in
Lambert, N. A., Prasad, P. D., and Ganapathy, V. (2009) GPR109A is a metabolic homeostasis: more friend than foe? Mol. Metab. 4,
G-protein-coupled receptor for the bacterial fermentation product 367–377
butyrate and functions as a tumor suppressor in colon. Cancer Res. 69, 54. Mielenz, M., Kuhla, B., and Hammon, H. M. (2013) Abundance of
2826–2832 adiponectin system and G-protein coupled receptor GPR109A mRNA
36. Ganji, S. H., Qin, S., Zhang, L., Kamanna, V. S., and Kashyap, M. L. in adipose tissue and liver of F2 offspring cows of Charolais 3 German
(2009) Niacin inhibits vascular oxidative stress, redox-sensitive genes, Holstein crosses that differ in body fat accumulation. J. Dairy Sci. 96,
and monocyte adhesion to human aortic endothelial cells. Athero- 278–289
sclerosis 202, 68–75 55. Li, X., Millar, J. S., Brownell, N., Briand, F., and Rader, D. J. (2010)
37. Gautam, J., Banskota, S., Shah, S., Jee, J. G., Kwon, E., Wang, Y., Kim, Modulation of HDL metabolism by the niacin receptor GPR109A in
D. Y., Chang, H. W., and Kim, J. A. (2018) 4-Hydroxynonenal-induced mouse hepatocytes. Biochem. Pharmacol. 80, 1450–1457
GPR109A (HCA2 receptor) activation elicits bipolar responses, Gai 56. Katsurada, A., Iritani, N., Fukuda, H., Matsumura, Y., Nishimoto, N.,
-mediated anti-inflammatory effects and Gbg -mediated cell death. Br. Noguchi, T., and Tanaka, T. (1990) Effects of nutrients and
J. Pharmacol. 175, 2581–2598 hormones on transcriptional and post-transcriptional regulation of
38. Graff, E. C., Fang, H., Wanders, D., and Judd, R. L. (2016) Anti- acetyl-CoA carboxylase in rat liver. Eur. J. Biochem. 190, 435–441
inflammatory effects of the hydroxycarboxylic acid receptor 2. Me- 57. Ruderman, N. B., Saha, A. K., and Kraegen, E. W. (2003) Minireview:
tabolism 65, 102–113 malonyl CoA, AMP-activated protein kinase, and adiposity. Endocri-
39. Singh, N., Gurav, A., Sivaprakasam, S., Brady, E., Padia, R., Shi, H., nology 144, 5166–5171
Thangaraju, M., Prasad, P. D., Manicassamy, S., Munn, D. H., Lee, 58. Titchenell, P. M., Chu, Q., Monks, B. R., and Birnbaum, M. J. (2015)
J. R., Offermanns, S., and Ganapathy, V. (2014) Activation of Hepatic insulin signalling is dispensable for suppression of glucose
Gpr109a, receptor for niacin and the commensal metabolite output by insulin in vivo. Nat. Commun. 6, 7078

4778 Vol. 33 April 2019 The FASEB Journal x www.fasebj.org YE ET AL.


59. Shi, Y., and Burn, P. (2004) Lipid metabolic enzymes: emerging drug 68. Besnard, P., Niot, I., Poirier, H., Clément, L., and Bernard, A. (2002)
targets for the treatment of obesity. Nat. Rev. Drug Discov. 3, 695–710 New insights into the fatty acid-binding protein (FABP) family in the
60. Rui, L. (2014) Energy metabolism in the liver. Compr. Physiol. 4, small intestine. Mol. Cell. Biochem. 239, 139–147
177–197 69. Garg, A., and Grundy, S. M. (1990) Nicotinic acid as therapy for
61. Kamanna, V. S., Ganji, S. H., and Kashyap, M. L. (2013) Recent dyslipidemia in non-insulin-dependent diabetes mellitus. JAMA 264,
advances in niacin and lipid metabolism. Curr. Opin. Lipidol. 24, 723–726
239–245 70. Goldberg, R. B., and Jacobson, T. A. (2008) Effects of niacin on
62. Cannon, B., and Nedergaard, J. (2004) Brown adipose tissue: function glucose control in patients with dyslipidemia. Mayo Clin. Proc. 83,
and physiological significance. Physiol. Rev. 84, 277–359 470–478
63. Gesta, S., Tseng, Y. H., and Kahn, C. R. (2007) Developmental origin 71. Chang, A. M., Smith, M. J., Galecki, A. T., Bloem, C. J., and Halter, J. B.
of fat: tracking obesity to its source. Cell 131, 242–256 (2006) Impaired beta-cell function in human aging: response to
64. Cohen, P., Levy, J. D., Zhang, Y., Frontini, A., Kolodin, D. P., Svensson,
nicotinic acid-induced insulin resistance. J. Clin. Endocrinol. Metab. 91,
K. J., Lo, J. C., Zeng, X., Ye, L., Khandekar, M. J., Wu, J., Gunawardana,
3303–3309
S. C., Banks, A. S., Camporez, J. P., Jurczak, M. J., Kajimura, S., Piston,
72. Wang, N., Guo, D. Y., Tian, X., Lin, H. P., Li, Y. P., Chen, S. J., Fu, Y. C.,
D. W., Mathis, D., Cinti, S., Shulman, G. I., Seale, P., and Spiegelman,
B. M. (2014) Ablation of PRDM16 and beige adipose causes metabolic Xu, W. C., and Wei, C. J. (2016) Niacin receptor GPR109A inhibits
dysfunction and a subcutaneous to visceral fat switch. Cell 156, insulin secretion and is down-regulated in type 2 diabetic islet beta-
304–316 cells. Gen. Comp. Endocrinol. 237, 98–108
65. Harms, M., and Seale, P. (2013) Brown and beige fat: development, 73. Li, H. M., Zhang, M., Xu, S. T., Li, D. Z., Zhu, L. Y., Peng, S. W., Chen,
function and therapeutic potential. Nat. Med. 19, 1252–1263 G. Q., Martin, P. M., Ganapathy, V., and Wei, C. J. (2011) Nicotinic
66. Altmann, S. W., Davis, H. R., Jr., Zhu, L. J., Yao, X., Hoos, L. M., acid inhibits glucose-stimulated insulin secretion via the G protein-
Tetzloff, G., Iyer, S. P., Maguire, M., Golovko, A., Zeng, M., Wang, L., coupled receptor PUMA-G in murine islet b cells. Pancreas 40,
Murgolo, N., and Graziano, M. P. (2004) Niemann-Pick C1 like 1 615–621
protein is critical for intestinal cholesterol absorption. Science 303, 74. Fulcher, G. R., Walker, M., Catalano, C., Agius, L., and Alberti, K. G.
1201–1204 (1992) Metabolic effects of suppression of nonesterified fatty acid
67. Hall, A. M., Wiczer, B. M., Herrmann, T., Stremmel, W., and Bernlohr, levels with acipimox in obese NIDDM subjects. Diabetes 41, 1400–1408
D. A. (2005) Enzymatic properties of purified murine fatty acid
transport protein 4 and analysis of acyl-CoA synthetase activities in Received for publication September 13, 2018.
tissues from FATP4 null mice. J. Biol. Chem. 280, 11948–11954 Accepted for publication December 3, 2018.

GPR109A REGULATES ENERGY HOMEOSTASIS 4779

You might also like