IDH1-dependent α-KG regulates brown fat differentiation and function by modulating histone methylation

IDH1-dependent α-KG regulates brown fat differentiation and function by modulating histone methylation

Journal Pre-proof IDH1-dependent α-KG regulates brown fat differentiation and function by modulating histone methylation Hyun Sup Kang, Jae Ho Lee, K...

14MB Sizes 1 Downloads 55 Views

Journal Pre-proof IDH1-dependent α-KG regulates brown fat differentiation and function by modulating histone methylation

Hyun Sup Kang, Jae Ho Lee, Kyoung-Jin Oh, Eun Woo Lee, Baek Soo Han, Kun-Young Park, Jae Myoung Suh, Jeong-Ki Min, Seung-Wook Chi, Sang Chul Lee, Kwang-Hee Bae, Won Kon Kim PII:

S0026-0495(20)30037-8

DOI:

https://doi.org/10.1016/j.metabol.2020.154173

Reference:

YMETA 154173

To appear in:

Metabolism

Received date:

14 November 2019

Accepted date:

4 February 2020

Please cite this article as: H.S. Kang, J.H. Lee, K.-J. Oh, et al., IDH1-dependent αKG regulates brown fat differentiation and function by modulating histone methylation, Metabolism(2020), https://doi.org/10.1016/j.metabol.2020.154173

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

© 2020 Published by Elsevier.

Journal Pre-proof

1 2

IDH1-dependent α-KG regulates brown fat differentiation and function

3

by modulating histone methylation

4 5

Hyun Sup Kang

6

Kun-Young Park c, Jae Myoung Suh c,d, Jeong-Ki Min a,b, Seung-Wook Chi a,b, Sang Chul Lee

7

a,b

a,b,1

, Jae Ho Lee

a,1

, Kyoung-Jin Oh

, Eun Woo Lee a, Baek Soo Han

a,b

,

oo

f

, Kwang-Hee Bae a,b,*, Won Kon Kim a,b,*

a

pr

8

9

a,b

Metabolic Regulation Research Center, Korea Research Institute of Bioscience and

Biotechnology (KRIBB), Daejeon 34141, Republic of Korea

11

b

12

Science and Technology (UST), Daejeon 34141, Republic of Korea

13

c

14

Republic of Korea

15

d

e-

10

Pr

Department of Functional Genomics, KRIBB School of Bioscience, Korea University of

al

Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141,

rn

Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea

Jo u

16

17

*Corresponding authors.

18

E-mail addresses: [email protected] (K.-H. Bae), [email protected] (W.K. Kim).

19

1

These two authors contributed equally to this work.

20

21

Abbreviations

22

IDH1, isocitrate dehydrogenase 1; α-KG, α-ketoglutarate; UCP1, uncoupling protein 1;

23

PPARγ, peroxisome proliferator-activated receptor-γ; PGC1α, PPARγ -coactivator protein 1-

24

alpha; PRDM16, PR-domain-containing 16; C/EBP, CCAAT-enhancer-binding protein. 1

Journal Pre-proof ABSTRACT

26

Objective. Brown adipocytes play important roles in the regulation of energy homeostasis by

27

uncoupling protein 1-mediated non-shivering thermogenesis. Recent studies suggest that

28

brown adipocytes as novel therapeutic targets for combating obesity and associated diseases,

29

such as type II diabetes. However, the molecular mechanisms underlying brown adipocyte

30

differentiation and function are not fully understood.

31

Methods. We employed previous findings obtained through proteomic studies performed to

32

assess proteins displaying altered levels during brown adipocyte differentiation. Here, we

33

performed assays to determine the functional significance of their altered levels during brown

34

adipogenesis and development.

35

Results. We identified isocitrate dehydrogenase 1 (IDH1) as upregulated during brown

36

adipocyte differentiation, with subsequent investigations revealing that ectopic expression of

37

IDH1 inhibited brown adipogenesis, whereas suppression of IDH1 levels promoted

38

differentiation of brown adipocytes. Additionally, Idh1 overexpression resulted in increased

39

levels of intracellular α-ketoglutarate (α-KG) and inhibited the expression of genes involved

40

in brown adipogenesis. Exogenous treatment with α-KG reduced brown adipogenesis during

41

the early phase of differentiation, and ChIP analysis revealed that IDH1-mediated α-KG

42

reduced trimethylation of histone H3 lysine 4 in the promoters of genes associated with

43

brown adipogenesis. Furthermore, administration of α-KG decreased adipogenic gene

44

expression by modulating histone methylation in brown adipose tissues of mice.

45

Conclusion. These results suggested that the IDH1–α-KG axis plays an important role in

46

regulating brown adipocyte differentiation and might represent a therapeutic target for

47

treating metabolic diseases.

48

Keywords: brown adipocyte differentiation, isocitrate dehydrogenase 1 (IDH1), α-

49

ketoglutarate (α-KG), histone modification

Jo u

rn

al

Pr

e-

pr

oo

f

25

2

Journal Pre-proof 50

1. Introduction

51

Obesity is a phenomenon primarily caused by sustained energy imbalance and represents a

52

major causal factor of various metabolic disorders [1, 2]. Due to the rapid increase in the

53

obese population and associated metabolic diseases, research into strategies for treating

54

obesity has become increasingly important. Adipose tissues are mainly composed of adipocytes that play critical roles in regulating

56

energy homeostasis. Mammals harbor two types of adipocytes, including white and brown

57

adipocytes. White adipocytes comprise a single, large lipid droplet located in the

58

subcutaneous and abdominal areas of the body where they store excess energy in the form of

59

triglycerides. Brown adipocytes are enriched with numerous mitochondria and mainly located

60

in the interscapular region in rodents and human infants. In contrast to white adipocytes,

61

brown adipocytes primarily consume energy to generate heat, with their thermogenic capacity

62

largely due to induction of the brown adipose tissue-specific protein uncoupling protein 1

63

(UCP1) located in the mitochondrial inner membrane [3]. UCP1 dissipates the mitochondrial

64

electrochemical gradient through proton leakage to induce uncoupled respiration [4, 5].

65

Therefore, the UCP1-mediated thermogenic functions of brown adipocytes are now regarded

66

as a therapeutic target for treating obesity [6].

Jo u

rn

al

Pr

e-

pr

oo

f

55

67

Brown adipocytes are derived from mesenchymal stem cells (MSCs), which are

68

multipotent stromal cells capable of differentiating into various cell types, such as adipocytes,

69

myocytes, chondrocytes, and osteoblasts, under specific stimuli [7]. Brown adipocyte

70

differentiation from MSCs involves two steps: 1) commitment to preadipocytes from MSCs

71

and 2) adipocyte differentiation into mature adipocytes [8, 9]. In particular, MSC

72

commitment to a brown adipocyte lineage is controlled by a variety of factors, including PR-

73

domain-containing 16 (PRDM16), bone morphogenetic protein 7 (BMP7), and early B cell

74

factor 2 (EBF2) [10-12]. In particular, PRDM16 coupled with CCAAT-enhancer-binding 3

Journal Pre-proof protein-β (C/EBPβ) is essential for the determination of brown fat from muscle-precursor

76

cells [13]. After lineage determination to brown preadipocytes, these cells sequentially

77

differentiate into mature brown adipocytes accompanied by the activities of multiple

78

transcriptional regulators, including peroxisome proliferator-activated receptor-γ (PPARγ),

79

C/EBPs, and PPARγ coactivator 1-alpha (PGC1α) [14, 15]. Specifically, PPARγ is a key

80

transcription factor involved in regulating brown adipocyte differentiation and the browning

81

of white adipose tissues [16]. Therefore, several transcriptional regulators are directly

82

involved in lineage determination and differentiation; however, the molecular mechanisms

83

that coordinate the commitment and differentiation of brown adipocytes remain incompletely

84

understood.

pr

oo

f

75

Histone modification plays a key role in the regulation of gene expression and

86

numerous cellular functions [17]. Among the various types of histone modification,

87

acetylation and methylation on lysine or arginine residues of histones H3 and H4 play

88

important biological roles, including transcriptional regulation [18, 19]. Gene expression is

89

differentially controlled depending on the site of histone methylation, with methylation of the

90

4th and 36th lysine residues of histone H3 responsible for general activation of transcription,

91

whereas methylation of the 9th and 27th lysine residues of histone H3 mediate gene

92

inactivation [19]. Moreover, methylation of these lysine residues can occur as mono-, di- and

93

trimethylation, which regulates genes in different ways [20]. Additionally, histone

94

methylation and demethylation play important roles in adipocyte differentiation [21] and

95

reportedly control adipocyte differentiation by regulating PPARγ expression [22].

96

Furthermore, a recent study reported the involvement of H3K27 methylation and acetylation

97

in the thermogenic program of brown adipocytes by regulating the expression of brown

98

adipocyte-specific genes, including Ucp1 and Pgc1a [23].

Jo u

rn

al

Pr

e-

85

4

Journal Pre-proof We previously performed proteomic analysis to screen changes in protein levels

100

during brown adipocyte differentiation [24] and identified isocitrate dehydrogenase 1 (IDH1)

101

as upregulated during differentiation of primary brown preadipocytes. IDH1 is an NADP+-

102

dependent metabolic enzyme responsible for converting isocitrate to α-ketoglutarate (α-KG)

103

[25, 26]. Although IDH2 localizes to the mitochondrial matrix, IDH1 localizes to the cytosol;

104

however, both IDH1/2 play essential roles in cellular metabolism, including the TCA cycle

105

[27]. In addition to its metabolic functions, IDH1 regulates gene expression through

106

epigenetic modification of histone proteins [28]. In this study, we investigated the functional

107

roles of IDH1 and its regulatory mechanisms associated with brown adipocyte differentiation.

Jo u

rn

al

Pr

e-

pr

oo

f

99

5

Journal Pre-proof 2. Materials and methods

109

2.1. Cell culture, brown adipocyte differentiation, and Oil-Red-O staining

110

An immortalized brown preadipocyte cell line was kindly provided by Dr. Shingo Kajimura

111

(UCSF, San Francisco, CA, USA). A brown preadipocyte cell line was obtained from the

112

interscapular brown adipose tissue of C57BL/6 mice at postnatal days 1–2 and isolated by

113

collagenase dispersion, as described previously [29]. These cells were grown in Dulbecco's

114

modified Eagle medium (DMEM) containing 1% antibiotics and 10% fetal bovine serum

115

(FBS) at 37C in a humidified atmosphere with 5% CO2. Primary brown preadipocytes were

116

isolated from the interscapular brown adipose tissues of 1- to 3-day old mice, as described

117

previously [30], and cultured in DMEM containing 1% antibiotics and 20% FBS (Gibco;

118

Invitrogen, Carlsbad, CA, USA) at 37°C in a humidified atmosphere with 5% CO2. For

119

brown adipocyte differentiation, cells were induced, as previously described [31]. The

120

C3H10T1/2 MSC line was provided by Dr. Jae Bum Kim (Seoul National University, Seoul,

121

South Korea) and grown in DMEM containing 1% antibiotics and 10% FBS at 37C. The

122

conditions used for adipogenic differentiation of C3H10T1/2 cells were previously described

123

[32].

Jo u

rn

al

Pr

e-

pr

oo

f

108

124

Lipid droplets of differentiated brown adipocytes were subjected to Oil-Red-O staining,

125

as described in our previous study [33]. Briefly, cultured cells were washed twice with

126

phosphate-buffered saline (PBS) and fixed for 30 min with 10% formaldehyde at room

127

temperature. Fixed cells were then washed with distilled water and stained for 30 min at room

128

temperature with 0.3% filtered Oil-Red-O solution in 60% isopropanol (Sigma-Aldrich, St.

129

Louis, MO, USA). The cells were then washed twice with distilled water, and micrographs

130

were obtained. For quantification analysis, Oil-Red-O staining dye was eluted, as previously

131

described [31].

132 6

Journal Pre-proof 2.2. Transduction using retroviral expression or knockdown vectors

134

To construct immortalized brown preadipocytes stably expressing FLAG-tagged mouse Idh1,

135

a retroviral infection system was used. For Idh1 expression, DNA encoding the FLAG-tagged

136

IDH1 was inserted into the pRetroX-IRES-ZsGreen1 vector (Clontech Laboratories,

137

Mountain View, CA, USA), as described previously [34]. For virus production, GP2-293 cells

138

were transfected using Lipofectamine 2000 (Gibco; Invitrogen), and infected cells were

139

selected using a FACSAria cell sorter (BD Biosciences, Franklin Lakes, NJ, USA) and

140

maintained in growth medium. The ectopic expression of IDH1 was determined by western

141

blot analysis and real-time polymerase chain reaction (PCR).

oo

f

133

To knockdown endogenous Idh1 expression, we used a retrovirus-mediated infection

143

system. Short-hairpin RNAs (shRNAs) were designed by selecting a target sequence for the

144

mouse Idh1 gene according to a previous report [35] and an RNA-interference target-

145

sequence selector (Clontech). The gene encoding shRNA against Idh1 was inserted into the

146

multi-cloning site of the pSIREN-RetroQ-DsRed vector (Clontech). The following gene-

147

specific sequences were used to successfully inhibit Idh1 expression: shIDH1-1, 5′-

148

GATCCGCTGCAGAGGCTTTAAAGATTCAAGAGATCTTTAAAGCCTCTGCAGCTTTTTTACGCGTG-3′ and 5′-

149

AATTCACGCGTAAAAAAGCTGCAGAGGCTTTAAAGATCTCTTGAATCTTTAAAGCCTCTGCAGCG-3′;shIDH1-2,

150

5′-GATCCGCACCATCCGAAACATTCTTTCAAGAGAAGAATGTTTCGGATGGTGCTTTTTTACGCGTG-3′ and 5′-

151

AATTCACGCGTAAAAAAGCACCATCCGAAACATTCTTCTCTTGAAAGAATGTTTCGGATGGTGCG;

152

shIDH1-3, 5′-GATCCGCATGCATATGGGGACCAATTTCAAGAGAATTGGTCCCCATATGCATGTTTTTTACGCGTG-

153

3′ and 5′-AATTCACGCGTAAAAAACATGCATATGGGGACCAATTC TCTTGAAATTGGTCCCCATATGCATGCG-3′.

154

Non-targeting control shRNA (scrambled; SCR) was provided by Clontech.

Jo u

rn

al

Pr

e-

pr

142

155 156

2.3. Quantitative reverse transcription (qRT)-PCR

7

and

Journal Pre-proof 157

Total RNAs were extracted from cultured cells using TRIzol reagent (Invitrogen) according

158

to manufacturer instructions, and cDNA was synthesized from total RNA using the reverse

159

transcriptase M-MLV and a random primer (Promega, Madison, WI, USA) according to

160

manufacturer instructions [36]. Amplified cDNA was analyzed by qRT-PCR using a SYBR

161

green PCR kit and each primer (Table S1). Gene-expression levels were normalized to that of

162

the TATA-binding protein (TBP) gene.

163

2.4. Chromatin immunoprecipitation (ChIP) assay

165

ChIP assays were performed, as previously described [23, 37]. For each immunoprecipitation

166

reaction, proteins in supernatants were immunoprecipitated with antibodies against histone

167

H3 trimethylated at lysine 4 (H3K4me3) and lysine 36 (H3K36me3; Abcam, Cambridge,

168

UK), respectively, with anti-rabbit IgG used as a negative control (GE Healthcare, Pittsburgh,

169

PA, USA). Precipitated DNA fragments were analyzed by real-time PCR using primers

170

against

171

CCCACTAGCAGCTCTTTGGA-3′

172

cAMP-CRE

173

AAAAGTAGGCTGGGCTGTCA-3′;

174

ATACACTGCCCTGTGTAAGG-3′ and 5′-CTGCTAGGTTGGCAAGGAAT-3′; pyruvate

175

dehydrogenase

176

GGGAGGTCTAGAGCCCCTAA-3′; and Wntless (Wls) 5′-CTGGCTGTGGCTTGTGTAAA-

177

3′ and 5′-GGACAAGAGGCAAAAGCAAC-3′.

Pr

e-

pr

oo

f

164

kinase

as

and

follows:

Ucp1

proximal

3

(Pdk3),

promoter,

5′-CTGTGGAGCAGCTCAAAGGT-3′;

5′-CAAAGCTGGCTTCAGTCACA-3′

Jo u

region,

promoters,

al

mouse

rn

relevant

Pparg

proximal

5′-

Pgc1a

and

5′-

promoter,

5′-

5′-TTCCTTAAAGCCCCGGTAAC-3′

and

5′-

178 179

2.5. Western blot

180

Cells were washed three times with ice-cold PBS and harvested in ice-cold NP-40 lysis buffer

181

(137 mM NaCl, 20 mM Tris-Cl, 1 mM EDTA, 10% glycerol, and 1% NP-40) containing a 8

Journal Pre-proof protease inhibitor and a phosphatase-inhibitor cocktail (Roche, Basel, Switzerland). Protein

183

concentrations were measured using a Bradford assay (Bio-Rad, Hercules, CA, USA).

184

Sodium dodecyl sulfate polyacrylamide gel electrophoresis and western blot analyses were

185

performed using standard protocols, as described previously [38]. The following primary

186

antibodies were used: IDH1 (Cell Signaling, Danvers, MA, USA; #3997), IDH2 (Cell

187

Signaling; #56439), UCP1 (Abcam; #ab10983), PGC1α (Invitrogen; #PA5-38021), PRDM16

188

(R&D Systems, Minneapolis, MN, USA; #AF6295), PPARγ (Cell Signaling; #2435), heat-

189

shock protein 90 (HSP90; Santa Cruz Biotechnology, Dallas, TX, USA; #sc-13119),

190

H3K4me3 (Abcam; #ab8580), H3K9me3 (Abcam; #ab8898), H3K27me3 (Abcam; #ab6002),

191

H3K36me3 (Abcam; #ab9050), Histone H3 (Abcam; #ab1791), Anti-5′ AMP-activated

192

protein kinase-alpha (AMPKα, Cell Signaling; #2532), and anti-phospho-AMPKα (Cell

193

Signaling; #2535). The specific signals were amplified by horseradish peroxidase-conjugated

194

secondary anti-rabbit, anti-mouse, or anti-sheep IgG antibody (Santa Cruz Biotechnology),

195

and were visualized using an enhanced chemiluminescence system (Fusion Solo S; Vilber

196

Lourmat, France). Relative amounts of each protein were quantified using ImageJ software.

rn

al

Pr

e-

pr

oo

f

182

Jo u

197 198

2.6. α-KG assay

199

The cell-membrane-permeable α-KG precursor dimethyl-2-ketoglutarate (dm-α-KG) was

200

purchased from Sigma-Aldrich [39]. Briefly, 1 mM dm-α-KG was used to treat immortalized

201

brown preadipocytes during differentiation, and intracellular α-KG concentrations were

202

analyzed using an α-KG assay kit (K677; BioVision, Milpitas, CA, USA) according to

203

manufacturer instructions. Similar to in vitro analysis, intracellular α-KG concentrations in

204

mouse tissues were measured using identical methods.

205 206

2.7. Animal experiments 9

Journal Pre-proof 207

All animal housing was in compliance and experiments were conducted in accordance with

208

the Korea Research Institute of Bioscience and Biotechnology (KRIBB) Institutional Animal

209

Care and Use Committee Guidelines. Mice were housed in a temperature- and humidity-

210

controlled, specific pathogen-free animal facility at 22℃ under a 12:12 hour light:dark cycle.

211

For studies in brown fat development, interscapular brown adipose tissues of mice from

212

embryonic day 18 and after birth from postnatal day 1 to 6 were carefully dissected and

213

analyzed by qRT-PCR and Western blotting (n = 2). For administration of α-KG in vivo, 8-

214

week-old

215

intraperitoneally with saline or 0.5, 1, and 2 g/kg of dm-α-KG (Sigma-Aldrich) for 3 hours (n

216

= 4). In addition, we sacrificed mice and dissected brown adipose tissues after 3 days of daily

217

intraperitoneal injections of 1 g/kg of dm-α-KG (n = 5). For cold exposure experiments, 11-

218

week-old C57BL/6 male mice were randomly divided into four groups: room temperature

219

(RT), cold exposure for 1, 3, and 7 days (n = 3). Mice as control group were all placed at RT

220

(22°C) for 7 days, while the cold groups were placed at 5°C for the above mentioned times.

mice

(KOATECH,

Pyeongtaek,

Korea)

f

male

were

injected

221

rn

al

Pr

e-

pr

oo

C57BL/6

2.8. Statistical analysis

223

All experiments were performed at least in triplicate. All data are presented as the mean ±

224

standard deviation (SD). Statistical significance of differences between two groups was

225

measured using the two-tailed Student's t test. For assessment between more than three

226

groups, we used one-way analysis of variance (ANOVA) with multiple comparisons. To

227

assess the relationship between two independent variables, two-way ANOVA with multiple

228

comparisons was used. One-way ANOVA was followed by Tukey's post hoc test, and two-

229

way ANOVA was followed by Sidak's multiple comparison test. Statistical analyses were

230

performed using GraphPad Prism (GraphPad Software, San Diego, CA, USA), and

231

differences were considered significant at p < 0.05.

Jo u

222

10

Journal Pre-proof 3. Results

233

3.1. IDH1 is upregulated during brown adipogenesis and brown adipose tissue

234

development

235

We previously performed proteomic analysis to identify differential levels of proteins during

236

brown adipocyte differentiation [24], eventually choosing IDH1 for further analysis. In the

237

present study, we found that IDH1 levels were clearly elevated in both immortalized brown

238

preadipocytes and primary brown preadipocytes during differentiation (Fig. 1A), whereas

239

IDH2 levels were unchanged during brown adipogenesis (Fig. 1A). Accordingly, Idh1 mRNA

240

levels were upregulated during brown adipogenesis along with upregulated expression of

241

brown adipogenic genes such as Ucp1 and Pgc1a (Fig. 1B). Next, we analyzed the

242

expression levels of IDH1 protein during brown fat development through late embryogenesis,

243

postnatal periods and adulthood. The levels of IDH1 and brown adipogenic proteins, UCP1

244

and PGC1α, increased from embryonic day 18 to postnatal day 6 when brown adipose tissue

245

became fully mature (Fig. 1C and D). These data show that IDH1 expression is upregulated

246

during brown fat adipogenesis both in vitro and in vivo.

oo

pr

e-

Pr

al

rn Jo u

247

f

232

248

3.2. Ectopic expression of IDH1 inhibits brown adipocyte differentiation

249

To investigate the functional role of IDH1 in brown adipocyte differentiation, we established

250

immortalized brown preadipocytes stably expressing Idh1 following retroviral infection.

251

After flow cytometric sorting of infected cells, stable expression was verified by fluorescence

252

microscopy (Fig. 2A), with ectopic expression of IDH1 determined by western blot (Fig. 2B).

253

The infected brown preadipocytes were then induced to differentiate into mature brown

254

adipocytes by culture in brown adipogenic induction medium for 6 days, followed by

255

examination of cellular lipid accumulation using Oil-Red-O staining on day 6 after

256

differentiation. The results showed that Idh1 overexpression decreased differentiation of 11

Journal Pre-proof 257

brown preadipocytes (Fig. 2C and D). Additionally, transcript levels of key genes involved in

258

brown adipogenesis, including Ucp1, Pgc1a, Prdm16, and Pparg, were significantly

259

suppressed upon IDH1 ectopic expression (Fig. 2E). Moreover, we consistently observed

260

attenuated UCP1 levels following differentiation in brown adipocytes ectopically expressing

261

IDH1 (Fig. 2B). These results strongly implied that IDH1 negatively regulated brown

262

adipocyte differentiation by suppressing the expression of genes involved in brown

263

adipogenesis.

oo

f

264

3.3. Idh1 knockdown promotes brown adipogenesis

266

To clarify the roles of IDH1 in regulating brown adipocyte differentiation, we generated

267

stable Idh1-knockdown brown preadipocytes using the pSIREN-RetroQ-DsRed vector

268

system (Fig. 3A). We verified the highly efficient knockdown of endogenous Idh1 expression

269

by shRNA IDH1-II according to qRT-PCR and western blot analyses (Fig. 3B) and observed

270

continuous suppression of Idh1 expression into the late phases of differentiation (Fig. 3C and

271

E). We then differentiated Idh1-knockdown immortalized brown preadipocytes into mature

272

brown adipocytes using a standard protocol for 6 days, followed by Oil-Red-O staining to

273

visually assess lipid accumulation. In contrast to Idh1 overexpression, Idh1 knockdown

274

promoted brown adipocyte differentiation relative to that observed in cells infected with

275

scrambled shRNA (Fig. 3D). Additionally, levels of UCP1, a key brown adipocyte protein,

276

were enhanced by Idh1 knockdown (Fig. 3E), with transcript levels of Ucp1, Pgc1a, Prdm16,

277

and Pparg also significantly upregulated in the presence of attenuated IDH1 levels (Fig. 3F).

Jo u

rn

al

Pr

e-

pr

265

278 279

3.4. α-KG inhibits brown adipocyte differentiation by acting at an early phase of

280

differentiation

12

Journal Pre-proof 281

Because IDH1 localizes to the cytosol and catalyzes the conversion of isocitrate to α-KG [25-

282

27], we investigated whether IDH1 regulates brown adipocyte differentiation through α-KG.

283

Measurement of intracellular α-KG levels during brown adipogenesis in cells ectopically

284

expressing IDH1 or Idh1-knockdown cells revealed fluctuations in α-KG levels according to

285

those of IDH1 during brown adipocyte differentiation (Fig. 4A), with elevated α-KG levels in

286

the presence of IDH1 and attenuated α-KG levels in Idh1-knockdown cells. These data

287

suggest that increases in α-KG levels might affect brown adipocyte differentiation. To examine the effects of α-KG on brown adipocyte differentiation, brown

289

preadipocytes were treated with cell-permeable dm-α-KG during differentiation, and its effect

290

on intracellular α-KG levels was determined. As shown in Fig. 4B, intracellular α-KG levels

291

were rapidly elevated following administration of dm-α-KG. We then added dm-α-KG to

292

culture medium at different time points during brown adipocyte differentiation, finding that

293

administration during the early phase (days 0–2) inhibited brown adipocyte differentiation,

294

whereas this activity during the middle (days 2–4) or terminal (days 4–6) stages did not affect

295

brown adipocyte differentiation or the expression of genes associated with brown

296

adipogenesis (Figs. 4C; S1A and B). Furthermore, we confirmed that protein and mRNA

297

levels of brown adipogenic markers, such as Ucp1, Pgc1a, Prdm16, and Pparg, were

298

consistently downregulated in mature brown adipocytes treated with α-KG for either 2 days

299

(days 0–2) or 6 days (days 0–6) (Fig. 4D and E). These results clearly implied that α-KG

300

affected the early phase of brown adipocyte differentiation by suppressing the expression of

301

key genes involved in brown adipogenesis, thereby inhibiting brown adipocyte differentiation.

Jo u

rn

al

Pr

e-

pr

oo

f

288

302 303

3.5. IDH1 negatively regulates brown adipocyte differentiation through α-KG

304

To clarify the α-KG-dependent effect of IDH1 on brown adipocyte differentiation, we

305

investigated whether supplementation of dm-α-KG of IDH1-depleted cells could rescue the 13

Journal Pre-proof 306

effects of Idh1 knockdown on brown adipocyte differentiation. Interestingly, the enhanced

307

brown adipocyte differentiation observed following Idh1 knockdown was potently

308

suppressed by administration of dm-α-KG (Fig. 5A). Additionally, qRT-PCR analysis

309

revealed that the elevated expression of genes associated with brown adipogenesis in Idh1-

310

knockdown cells was downregulated to control levels following dm-α-KG administration

311

(Fig. 5B). These results suggested that IDH1-mediated upregulation of α-KG levels is crucial

312

for regulating brown adipogenic genes and brown adipocyte differentiation. We then examined the effect of treatment with α-KG during brown adipocyte

314

differentiation in Idh1-overexpressing cells. Compared with the significant repression of

315

brown adipocyte differentiation observed following dm-α-KG administration or IDH1 ectopic

316

expression, Idh1-overexpressing cells administered dm-α-KG showed no additive effect on

317

inhibiting brown adipocyte differentiation (Fig. 5C), with similarly attenuated levels of

318

brown adipogenic transcripts also observed according to cell-treatment status (Fig. 5D).

319

These data suggested that IDH1-mediated α-KG level or exogenous dm-α-KG alone might be

320

sufficient to inhibit the expression of brown adipogenic genes and brown adipocyte

321

differentiation.

oo

pr

e-

Pr

al

rn Jo u

322

f

313

323

3.6. Brown adipocyte differentiation is regulated by α-KG-mediated alteration of

324

histone methylation

325

Previous studies report that IDH1 not only plays roles in metabolic pathways but also

326

promotes epigenetic reprogramming through α-KG [28, 39], which regulates gene expression

327

by activating several histone demethylases [40]. Therefore, we hypothesized that IDH1-

328

mediated α-KG affects brown adipocyte differentiation by modulating epigenetic regulation

329

of gene expression. First, we monitored histone-methylation patterns during brown adipocyte

330

differentiation by western blot using specific antibodies, finding that H3K4me3 and 14

Journal Pre-proof H3K36me3 (but not H3K9me3 and H3K27me3 levels) increased during the first 2 days of

332

brown adipogenesis (Fig. 6A). Since H3K4me3 and H3K36me3 represent active chromatin

333

complexes, we speculated that this might be related to the induction of adipogenic gene

334

expression [41, 42]. As expected, mRNA levels of brown adipogenic genes were elevated at

335

48-h post-induction of differentiation and attenuated in the presence of α-KG (Fig. 6B). We

336

then examined whether IDH1 and/or α-KG affect levels of histone methylation, finding that

337

H3K4me3 and H3K36me3 levels were reduced by treatment with α-KG or IDH1 ectopic

338

expression (Fig. 6C), with further decreases in these levels observed in α-KG-treated Idh1-

339

overexpressing cells (Fig. 6C). By contrast, attenuated IDH1 levels resulted in elevated

340

H3K4me3 and H3K36me3 levels presumably by reducing α-KG levels (Figs. 4A and 6D).

341

Additionally, replenishment of the α-KG level to that present in IDH1-depleted cells rescued

342

H3K4me3 and H3K36me3 levels (Fig. 6D). These results suggested that IDH1 increased

343

intracellular α-KG levels and subsequently suppressed specific histone methylation

344

(H3K4me3 and H3K36me3) to inhibit brown adipocyte differentiation.

al

Pr

e-

pr

oo

f

331

Because histone methylation in the promoters of genes associated with brown adipocyte

346

differentiation is associated with transcriptional regulation of each gene [23], we evaluated

347

the effect of IDH1 and/or α-KG on the pattern of histone methylation in the promoter regions

348

of Ucp1, Pgc1a, and Pparg using ChIP analysis. We found that H3K4me3 levels at these

349

promoters were significantly reduced in cells exhibiting IDH1 ectopic expression or

350

undergoing dm-α-KG treatment (Fig. 6E), whereas they were suppressed in Idh1-knockdown

351

and further attenuated to control levels by additional dm-α-KG treatment (Fig. 6F).

352

Interestingly, we did not observed changes in H3K36me3 levels in these promoters,

353

regardless of dm-α-KG treatment or IDH1 status, indicating that H3K36me3 is not involved

354

in the regulation of brown adipogenic genes during brown adipocyte differentiation (Fig. 6E

355

and F). Additionally, dm-α-KG treatment did not change levels of H3K4me3 and H3K36me3

Jo u

rn

345

15

Journal Pre-proof 356

in Prdm16-promoter regions (Fig. S2). These results suggested that IDH1-induced α-KG

357

specifically suppressed H3K4me3 in the promoters of brown adipogenic genes, thereby

358

preventing brown adipocyte differentiation. We then investigated whether IDH1 and α-KG also modulate histone methylation in the

360

promoters of white adipocyte-specific genes. ChIP analysis of the promoter regions of two

361

white adipocyte-specific genes, Wls and Pdk3 [37], indicated no significant changes to

362

histone modifications in these promoter regions or their transcript levels according to IDH1

363

status or α-KG treatment (Fig. S3A and B). These data strongly suggested that IDH1- and α-

364

KG-mediated fine-tuning of H3K4me3 levels in the promoter regions of brown adipogenic

365

genes during the early phase of differentiation is critical for brown adipocyte differentiation.

pr

oo

f

359

e-

366

3.7. α-KG administration decreased brown adipogenic gene expression in vivo

368

It has been demonstrated that brown adipogenic factors, such as UCP1 and PGC1α, are

369

required for the maintenance as well as the differentiation of brown adipocytes [43]. To

370

further investigate the effects of α-KG on maintenance and function of brown fat in vivo,

371

mice were injected intraperitoneally with α-KG. The increase of α-KG level in brown fat was

372

dependent on the concentration of administered α-KG, indicating the efficient delivery of

373

exogenous α-KG to brown adipose tissues (Fig. 7A). Since intracellular α-KG concentrations

374

were saturated by injection of 1 g/kg α-KG, we examined adipogenic gene expression after

375

the injection of 1 g/kg α-KG. Interestingly, exogenous α-KG decreased the mRNA levels of

376

the brown adipocyte-specific genes, such as Ucp1 and Pgc1a, in brown adipose tissues (Fig.

377

7B). In addition, α-KG suppressed the protein levels of brown adipogenic markers in brown

378

fat tissues (Fig. 7C). Moreover, we examined whether α-KG was accompanied by a change in

379

histone methylation in vivo. Similar to in vitro data, α-KG administration altered histone

380

methylation patterns, particularly H3K4me3 levels, in brown adipose tissues (Fig. 7D). In

Jo u

rn

al

Pr

367

16

Journal Pre-proof 381

accordance with the α-KG-mediated reduction of brown adipocyte-specific gene expression,

382

H3K4me3 levels at the promoters of these genes, including Ucp1 and Pgc1a, were decreased

383

in brown adipose tissues of α-KG-administered mice (Fig. 7E). On the other hand,

384

H3K36me3 occupancy at these promoters was not altered by α-KG supplementation in brown

385

adipose tissues (Fig. 7E). These data imply that α-KG may not only regulate brown

386

adipogenesis but also mature brown fat physiology and function.

387

3.8. AMPK activation prevents differentiation of brown preadipocytes

389

A previous study suggests that activation of the AMPK–α-KG axis increases C3H10T1/2

390

MSC differentiation into brown adipocytes [44]. To evaluate the role(s) of AMPK in brown

391

adipogenesis, we examined the differentiation capacity of brown preadipocytes following

392

treatment with the AMPK activator metformin. As shown in Fig. 8A, metformin efficiently

393

activated AMPK signaling based on elevated levels of phosphorylated AMPK in brown

394

preadipocytes. Consistent with a previous report [44], activation of AMPK enhanced

395

intracellular α-KG levels during brown adipocyte differentiation (Fig. 8B). Additionally,

396

metformin suppressed brown adipogenesis, lipid accumulation, as well as the expression of

397

brown adipogenic genes, including Ucp1, Pgc1a, and Pparg (Figs. 8C–E and S4A). These

398

results suggested that metformin inhibited brown adipogenesis via AMPK-mediated

399

alteration of α-KG level. Subsequent investigation of the effect of α-KG level on C3H10T1/2

400

MSC differentiation to brown adipocytes indicated that no change in differentiation or the

401

expression of brown adipogenic genes during this process (Figs. 8F–G and S4B).

Jo u

rn

al

Pr

e-

pr

oo

f

388

402 403

3.9. IDH1 and α-KG is regulated by cold-induced thermogenic activation in brown

404

adipose tissues

17

Journal Pre-proof Brown adipose tissue is a thermogenic organ that protects the body from cold exposure via

406

dissipating energy as heat [45]. Moreover, it has been demonstrated that TCA cycle

407

intermediates, especially succinate, regulate the activation of brown fat thermogenesis [46].

408

To elucidate whether IDH1 and α-KG might be involved in brown fat thermogenesis, we

409

analyzed the levels of IDH1 and α-KG in brown adipose tissues upon cold exposure. In

410

brown adipose tissues, the mRNA levels of Idh1 and thermogenic genes, Ucp1 and Pgc1a,

411

were increased in a manner dependent on the duration of cold exposure (Fig. 9A). In addition,

412

the level of IDH1 protein was upregulated under conditions of thermogenic activation by cold

413

challenge (Fig. 9B). Consistent with cold-induced IDH1 expression, the intracellular α-KG

414

levels in brown adipose tissues were elevated by cold exposure (Fig. 9C). Furthermore, cold

415

challenge modulated histone methylation patterns, especially induction of H3K4me3 levels

416

(Fig. 9D). These data suggested that the IDH1–α-KG axis may regulate in vivo brown fat

417

thermogenesis under cold exposure conditions.

Jo u

rn

al

Pr

e-

pr

oo

f

405

18

Journal Pre-proof 4. Discussion

419

Brown adipocytes are widely accepted as an optimal target for creating strategies to

420

overcome obesity and its related diseases; therefore, investigating brown adipocyte formation

421

represents an active research area. To identify novel factors involved in brown adipocyte

422

differentiation, we previously screened proteins exhibiting differential levels during brown

423

adipocyte differentiation using two-dimensional gel electrophoresis proteomics [24], which

424

identified IDH1 in primary brown adipocytes, as well as in immortalized brown adipocytes.

425

Moreover, the expression of IDH1 was increased during the development of brown adipose

426

tissues. On the other hand, the level of IDH1 in brown fat in 12-week-old mice was lower

427

compared to the perinatal periods. This expression pattern may reflect the requirement of

428

IDH1 when brown fat thermogenic demand is highest, such as during early postnatal stages

429

of life. IDH1 plays an important role in maintaining metabolic energy homeostasis by

430

converting isocitrate to α-KG [25-27]. Previous studies focused on the metabolic function of

431

IDH1 and its critical role of mutated IDH1 in cancer [47]; however, little is known about the

432

role of IDH1 in adipocyte differentiation and function. Therefore, we investigated the

433

functional roles and regulatory mechanisms of IDH1 in brown adipocyte metabolism.

Jo u

rn

al

Pr

e-

pr

oo

f

418

434

We found that ectopic expression of IDH1 in brown preadipocytes significantly

435

inhibited brown adipocyte differentiation, whereas knockdown of endogenous Idh1

436

expression increased brown adipocyte differentiation. Moreover, the presence of IDH1

437

increased α-KG levels, and the addition of exogenous α-KG suppressed brown adipocyte

438

differentiation at the early stage (days 0–2). These results suggested IDH1 as a potent

439

negative regulator of brown adipocyte differentiation at the early stage of differentiation by

440

modulating α-KG level.

441

Interestingly, the expression of IDH1, a negative regulator of brown adipogenesis,

442

increases during brown adipocyte differentiation indicating that IDH1 may play a role in fine19

Journal Pre-proof tuning brown adipocyte differentiation and function. In this regard, a previous study

444

demonstrated that Twist-1, which is induced upon PPARδ activation during brown

445

adipogenesis, inhibits Pgc1α expression in a negative-feedback regulatory loop to tightly

446

modulate PGC1α-controlled brown fat metabolism [48]. In addition, orphan nuclear receptor

447

NR4A, whose expression is upregulated during 3T3-L1 adipogenesis, serves as a potent

448

negative regulator of differentiation by inhibiting mitotic clonal expansion of 3T3-L1

449

preadipocytes [49]. In this study, multiple lines of in vitro and in vivo data support the idea

450

that the IDH1–α-KG axis functions as a negative-feedback regulator to fine-tune brown fat

451

differentiation and function.

oo

f

443

Since IDH1 catalyzes the transformation of isocitrate to α-KG, we focused on α-KG in

453

order to identify the mechanism by which IDH1 regulates brown adipogenic gene expression.

454

α-KG is an important cofactor of ten-eleven translocation dioxygenase, which regulates

455

histone methylation [50], and induces histone demethylation through activation of lysine

456

demethylases (KDMs) [40, 51]. Accordingly, decreased α-KG levels via reduced IDH1

457

activity increases histone-methylation levels [28]. Among various histone proteins and their

458

residues, H3K4 and H3K36 are critical for gene-promoter activation, with methylation of

459

H3K4 and H3K36 activating the transcription of certain gene subsets [21]. Moreover,

460

previous reports indicate that methylation of H3K4 and H3K36 regulates adipogenic gene

461

activity and expression [52, 53]. In particular, promoter methylation at H3K4 by mixed-

462

lineage leukemia 3 (also known as lysine N-methyltransferase 2C) activates adipocyte

463

differentiation [54]. Furthermore, demethylation of H3K4 by lysine-specific histone

464

demethylase 1A differentially controls brown adipose-specific versus white adipose-specific

465

gene expression [37]. Based on these reports, we propose that IDH1-dependent α-KG

466

regulates brown adipocyte-related gene expression through modulation of the histone-

467

methylation pattern in their respective promoters. Interestingly, in the present study, we

Jo u

rn

al

Pr

e-

pr

452

20

Journal Pre-proof 468

observed elevated H3K4me3 and H3K36me3 levels during the early stage of brown

469

adipocyte differentiation, and IDH1-mediated α-KG levels suppressed H3K4me3 levels in the

470

promoters of brown adipogenic genes. In agreement with in vitro data, in vivo administration

471

of α-KG reduced the expression of brown adipogenic genes in brown adipose tissues, which

472

was accompanied by decreased H3K4me3 levels at brown adipogenic gene promoters. These

473

results suggested that the IDH1–α-KG axis is important for brown adipocyte differentiation

474

and function dependent upon brown adipogenic gene expression. Brown adipocytes play an essential role in thermogenesis via β-adrenergic-dependent

476

and -independent pathways under cold conditions [45]. Recently, it has been demonstrated

477

that succinate, a TCA intermediate, is required for brown fat thermogenesis upon cold

478

challenge [46]. In this study, we observed an increase in the levels of IDH1 and α-KG, along

479

with those of UCP1 and PGC1α in brown adipose tissues by cold exposure. In addition, cold

480

exposure was also accompanied by changes in histone methylation, especially H3K4me3

481

levels, in brown adipose tissues. Unexpectedly, administration of CL-316,243, a β-adrenergic

482

agonist, did not change the level of IDH1 (Fig. S5). It is possible that cold exposure could

483

regulate the levels of IDH1 and α-KG by various factors other than β-adrenergic signaling.

484

These results are consistent with the notion that the IDH1–α-KG axis may play a key role in

485

thermogenesis of brown adipose tissues. Nevertheless, further studies are needed to elucidate

486

the effects of IDH1 and α-KG on the regulation of brown fat thermogenesis.

Jo u

rn

al

Pr

e-

pr

oo

f

475

487

By contrast, IDH1 reportedly promotes white adipocyte differentiation and lipid

488

synthesis via NADPH production [55]. In agreement with this report, we found that IDH1

489

was upregulated during differentiation of 3T3-L1 white adipocytes (Fig. S6A). Additionally,

490

Idh1 overexpression in 3T3-L1 cells promoted adipocyte differentiation, whereas Idh1

491

knockdown suppressed adipogenesis (Fig. S6B and C). In contrast to brown adipocytes,

492

H3K4me3 occupancy in the Pparg promoter was not altered by Idh1 overexpression and 21

Journal Pre-proof reduced by Idh1 knockdown in 3T3-L1 adipocytes (Fig. S6B and C). Notably, only

494

enrichment of H3K4me3 at brown adipocyte-specific promoters, but not at white adipocyte-

495

specific promoters, was attenuated by Idh1 overexpression or α-KG treatment (Fig. S3B).

496

Furthermore, differentiation of 3T3-L1 adipocytes was increased by treatment with α-KG and

497

accompanied by upregulated expression of adipogenic genes, including Pparg, aP2, and

498

adiponectin (Fig. S7A and B). These data suggest that IDH1 might differentially regulate

499

adipogenesis of brown and white adipocytes by modulating histone methylation in a cell-

500

type-selective manner. However, it remains unclear why IDH1/α-KG plays an opposing role

501

between white and brown adipocyte differentiation; therefore, extensive studies are warranted

502

to elucidate the specific role and mechanism of IDH1 in white and brown fat development

503

using brown adipocyte-specific Idh1-knockout animal models.

e-

pr

oo

f

493

A recent report indicated that AMPK-mediated α-KG promotes brown adipogenesis in

505

MSCs [44], demonstrating that ablation of AMPKα1 reduces α-KG levels and impairs

506

development of brown adipose tissues. Unexpectedly, our study revealed that the IDH1–α-

507

KG axis inhibits brown adipocyte differentiation; therefore, we suggest that α-KG might be

508

required for committing progenitor cells to the brown adipocyte lineage while possibly

509

inhibiting the differentiation of brown preadipocytes, which are already adipocyte

510

determinant. Generally, brown adipocyte differentiation comprises commitment to the brown

511

adipocyte lineage by MSCs and differentiation from preadipocytes to mature adipocytes [8,

512

9]. MSCs can differentiate into various cell types, including adipocytes, myocytes,

513

chondrocytes, and osteoblasts [7], with adipocytes requiring regulators, such as PRDM16,

514

BMP7, and EBF2, for commitment to the brown adipocyte lineage [10-12]. Moreover, α-KG

515

maintains the pluripotency of embryonic stem cells (ESCs) and inhibits the differentiation of

516

naive-state ESCs via epigenetic regulation [56], whereas α-KG also promotes early stage

517

neuroectodermal differentiation of pluripotent stem cells [57]. Therefore, α-KG is potentially

Jo u

rn

al

Pr

504

22

Journal Pre-proof involved in self-renewal and/or differentiation depending on the stage of cellular maturity,

519

suggesting that α-KG might control the brown adipogenic process in a stage-selective manner.

520

A previous study suggested stage-specific functions for pre-B-cell leukemia transcription

521

factor-1 in the control of adipocyte development by promoting the adipocyte-progenitor step

522

of ESCs while preventing adipocyte differentiation [58]. Additionally, dexamethasone is an

523

essential inducer of the early stage of adipocyte differentiation but displays anti-adipogenic

524

effects at terminal stages of adipogenesis, indicating time-dependent roles for hormones or

525

metabolites in adipogenesis [59]. Therefore, it is possible that α-KG in MSCs can drive

526

differentiation into the brown adipocyte lineage through DNA demethylation in the Prdm16

527

promoter, whereas α-KG in brown preadipocytes suppresses adipogenesis by inhibiting

528

adipogenic gene expression through histone modification.

e-

pr

oo

f

518

AMPK suppresses the differentiation of 3T3-L1 white adipocytes by modulating the

530

Akt–mammalian target of rapamycin, WNT/β-catenin, and extracellular signal-regulated

531

kinase pathways [60, 61]; however, little is known about the role of AMPK in brown

532

adipogenesis. A previous report showed that daily treatment with the AMPK activator 5-

533

aminoimidazole-4-carboxamide ribonucleotide (AICAR) is toxic to brown adipocytes by

534

decreasing intracellular pH [62]. To clarify the roles of AMPK in brown adipogenesis, we

535

examined the adipogenic capacity of the AMPK activator metformin following administration

536

to brown preadipocytes during differentiation. We found that metformin increased

537

intracellular α-KG levels and inhibited brown adipocyte differentiation, suggesting that

538

AMPK might inhibit differentiation of brown preadipocytes through elevated α-KG

539

production. Additionally, metformin also plays an AMPK-independent role in inhibiting

540

respiratory chain complex I [63]; therefore, it is possible that metformin suppresses brown

541

adipogenesis by regulating the mitochondrial respiratory chain.

Jo u

rn

al

Pr

529

23

Journal Pre-proof Mitochondrial respiration and lipogenesis are important metabolic pathways not only to

543

adipocyte function but also for cancer-cell survival [35]. PPARγ is a master regulator of

544

adipocyte differentiation and also controls glucose and lipid metabolism in cancer cells [64].

545

Additionally, PPARγ agonists can either promote or suppress tumorigenesis, tumor

546

progression, and metastasis in various tumor types [65]. Moreover, PGC1α plays an

547

important role in mitochondrial biogenesis and oxidative phosphorylation in cancer cells to

548

promote rapid proliferation and metastasis [66]. Therefore, further investigation is needed to

549

determine the roles of IDH1-mediated PPARγ–PGC1α regulatory pathways for cancer

550

metabolism. In the previous decade, gain-of-function mutations in IDH1/2 in cancers, and

551

especially glioma, were extensively studied [28]. Mutation of IDH1 at R132 results in the

552

production of 2-hydroxyglutaric acid (2-HG) rather than α-KG, which rewires cellular

553

metabolism to support tumorigenesis and cancer progression [67]. Furthermore, 2-HG

554

produced by mutant IDH1/2 inhibits the H3K9 demethylase KDM4C, thereby preventing the

555

differentiation of adipocytes, as well as astrocytes [28]. Therefore, IDH1, as well as mutant

556

IDH1/2, might be involved in brown adipocyte differentiation via α-KG- or 2-HG-mediated

557

histone modification, and this pathway might represent a therapeutic target for metabolic

558

diseases, including obesity and cancer.

Jo u

rn

al

Pr

e-

pr

oo

f

542

559

Taken together, we demonstrated that the IDH1–α-KG axis inhibits brown adipocyte

560

differentiation through histone modification-mediated transcriptional control of brown

561

adipogenic genes. Our results identified the molecular pathway associated with stage-

562

dependent IDH1-mediated α-KG regulation of brown adipocyte differentiation. Moreover, the

563

data suggest that IDH1–α-KG might inversely modulate adipogenesis of brown and white

564

adipocytes. Further studies are required to understand the underlying mechanisms involved in

565

the cell-type-selective opposite effects of IDH1 on adipogenesis. These findings suggest that

24

Journal Pre-proof IDH1 might represent a therapeutic target for treating obesity and metabolic diseases by

567

controlling adipocyte differentiation.

Jo u

rn

al

Pr

e-

pr

oo

f

566

25

Journal Pre-proof 5. Conclusions

569

In summary, we demonstrated that IDH1-mediated α-KG negatively regulated brown

570

adipocyte differentiation. Mechanistically, the IDH1–α-KG axis reduced H3K4me3 levels in

571

the promoters of brown adipogenic genes, which was accompanied by their decreased

572

expression. Moreover, administration of α-KG decreased adipogenic gene expression in

573

brown adipose tissues of mice, which were concomitant with alterations in histone

574

methylation. These findings suggest IDH1 is a key regulator of brown fat differentiation and

575

function. Taken together, the IDH1–α-KG axis might represent a potential therapeutic target

576

for ameliorating metabolic syndrome.

Jo u

rn

al

Pr

e-

pr

oo

f

568

26

Journal Pre-proof 577

Author contributions

578

HSK, JHL, K-JO, EWL, SCL, K-HB, and WKK conceived the study design, and data

579

interpretation. HSK, JHL, K-JO, EWL, K-YP, and JMS performed experiments. HSK, JHL,

580

JMS, K-HB, and WKK contributed to writing the manuscript. K-JO, EWL, BSH, J-KM, S-

581

WC, and SCL critically reviewed the study and gave the necessary suggestions. All authors

582

read and approved the final version of the manuscript.

oo

f

583

Acknowledgments

585

We thank Professor Jae Bum Kim at the Seoul National University for providing the

586

C3H10T1/2 cells. Additionally, we thank Dr. Hee Jun Cho at the Korea Research Institute of

587

Bioscience and Biotechnology for sharing the AMPK activator metformin. We also thank

588

Min Wook Kim for proofreading the manuscript. This work was supported by grants from the

589

KRIBB and the Research Program (grants 2017M3A9C4065954, 2015M3A9D7029882,

590

2017R1E1A1A01074745, and 2016R1C1B2013430) through the National Research Foundation

591

of Korea.

e-

Pr

al

rn Jo u

592

pr

584

593

Conflict of interest

594

The authors have no conflicts of interest and declare no competing financial interests.

27

Journal Pre-proof 595

References

596

[1] Guh DP, Zhang W, Bansback N, Amarsi Z, Birmingham CL, Anis AH. The incidence of co-

597

morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC

598

Public Health. 2009;9:88.

599

[2] Kopelman PG. Obesity as a medical problem. Nature. 2000;404:635-43.

600

[3] Park A, Kim WK, Bae KH. Distinction of white, beige and brown adipocytes derived from

607 608 609 610 611 612 613 614 615

f oo

606

Mol Cell Biol. 2005;6:248-61.

[6] Boss O, Farmer SR. Recruitment of brown adipose tissue as a therapy for obesity-associated diseases. Front Endocrinol (Lausanne). 2012;3:14.

pr

605

[5] Krauss S, Zhang CY, Lowell BB. The mitochondrial uncoupling-protein homologues. Nat Rev

[7] Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143-7.

e-

604

biology of mitochondrial uncoupling proteins. Diabetes. 2004;53 Suppl 1:S130-5.

[8] Cristancho AG, Lazar MA. Forming functional fat: a growing understanding of adipocyte differentiation. Nat Rev Mol Cell Biol. 2011;12:722-34.

Pr

603

[4] Rousset S, Alves-Guerra MC, Mozo J, Miroux B, Cassard-Doulcier AM, Bouillaud F, et al. The

[9] Kajimura S, Seale P, Spiegelman BM. Transcriptional control of brown fat development. Cell Metab. 2010;11:257-62.

[10] Seale P, Kajimura S, Yang W, Chin S, Rohas LM, Uldry M, et al. Transcriptional control of

al

602

mesenchymal stem cells. World J Stem Cells. 2014;6:33-42.

brown fat determination by PRDM16. Cell Metab. 2007;6:38-54.

rn

601

[11] Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, Taniguchi CM, et al. New role of

617

bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature.

618

2008;454:1000-4.

619 620 621 622 623 624 625 626 627 628 629 630

Jo u

616

[12] Rajakumari S, Wu J, Ishibashi J, Lim HW, Giang AH, Won KJ, et al. EBF2 determines and maintains brown adipocyte identity. Cell Metab. 2013;17:562-74. [13] Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;454:961-7. [14] Tontonoz P, Spiegelman BM. Fat and beyond: the diverse biology of PPARgamma. Annu Rev Biochem. 2008;77:289-312. [15] Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell. 1998;92:829-39. [16] Ohno H, Shinoda K, Spiegelman BM, Kajimura S. PPARgamma agonists induce a white-tobrown fat conversion through stabilization of PRDM16 protein. Cell Metab. 2012;15:395-404. [17] Wolffe AP. Transcriptional regulation in the context of chromatin structure. Essays Biochem. 2001;37:45-57. 28

Journal Pre-proof 631 632 633 634 635 636 637 638 639 640

[18] Bedford MT, Richard S. Arginine methylation an emerging regulator of protein function. Mol Cell. 2005;18:263-72. [19] Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol. 2005;6:838-49. [20] Zhang Y, Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 2001;15:2343-60. [21] Okamura M, Inagaki T, Tanaka T, Sakai J. Role of histone methylation and demethylation in adipogenesis and obesity. Organogenesis. 2010;6:24-32. [22] Wang L, Xu S, Lee JE, Baldridge A, Grullon S, Peng W, et al. Histone H3K9 methyltransferase G9a represses PPARgamma expression and adipogenesis. EMBO J. 2013;32:45-59. [23] Li F, Wu R, Cui X, Zha L, Yu L, Shi H, et al. Histone Deacetylase 1 (HDAC1) Negatively

642

Regulates Thermogenic Program in Brown Adipocytes via Coordinated Regulation of Histone

643

H3 Lysine 27 (H3K27) Deacetylation and Methylation. J Biol Chem. 2016;291:4523-36.

647 648 649

oo

pr

646

proteome during the differentiation of brown preadipocytes. J Proteomics. 2013;94:327-36.

e-

645

[24] Kamal AH, Kim WK, Cho K, Park A, Min JK, Han BS, et al. Investigation of adipocyte

[25] D AA, Jr., Haft DE. An Alternate Pathway of Alpha-Ketoglutarate Catabolism in the Isolated, Perfused Rat Liver. I. Studies with Dl-Glutamate-2- and -5-14c. J Biol Chem. 1965;240:613-7.

Pr

644

f

641

[26] Ernster L, Navazio F. The cytoplasmic distribution of isocitric dehydrogenases. Exp Cell Res. 1956;11:483-6.

[27] Xu X, Zhao J, Xu Z, Peng B, Huang Q, Arnold E, et al. Structures of human cytosolic NADP-

651

dependent isocitrate dehydrogenase reveal a novel self-regulatory mechanism of activity. J Biol

652

Chem. 2004;279:33946-57.

rn

al

650

[28] Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, et al. IDH mutation impairs

654

histone demethylation and results in a block to cell differentiation. Nature. 2012;483:474-8.

655

[29] Ohno H, Shinoda K, Ohyama K, Sharp LZ, Kajimura S. EHMT1 controls brown adipose cell fate

656

Jo u

653

and thermogenesis through the PRDM16 complex. Nature. 2013;504:163-7.

657

[30] Kim WK, Choi HR, Park SG, Ko Y, Bae KH, Lee SC. Myostatin inhibits brown adipocyte

658

differentiation via regulation of Smad3-mediated beta-catenin stabilization. Int J Biochem Cell

659

Biol. 2012;44:327-34.

660

[31] Kim WK, Oh KJ, Choi HR, Park A, Han BS, Chi SW, et al. MAP kinase phosphatase 3 inhibits

661

brown adipocyte differentiation via regulation of Erk phosphorylation. Mol Cell Endocrinol.

662

2015;416:70-6.

663

[32] McDonald ME, Li C, Bian H, Smith BD, Layne MD, Farmer SR. Myocardin-related

664

transcription factor A regulates conversion of progenitors to beige adipocytes. Cell.

665

2015;160:105-18.

29

Journal Pre-proof 666

[33] Son MJ, Kim WK, Park A, Oh KJ, Kim JH, Han BS, et al. Set7/9, a methyltransferase, regulates

667

the thermogenic program during brown adipocyte differentiation through the modulation of p53

668

acetylation. Mol Cell Endocrinol. 2016;431:46-53.

669

[34] Lee DS, Choi H, Han BS, Kim WK, Lee SC, Oh KJ, et al. c-Jun regulates adipocyte

670

differentiation via the KLF15-mediated mode. Biochem Biophys Res Commun. 2016;469:552-8.

671

[35] Nam WS, Park KM, Park JW. RNA interference targeting cytosolic NADP(+)-dependent

672

isocitrate dehydrogenase exerts anti-obesity effect in vitro and in vivo. Biochim Biophys Acta.

673

2012;1822:1181-8. [36] Kim J, Kim WK, Oh KJ, Lee EW, Han BS, Lee SC, et al. Protein tyrosine phosphatase, receptor

675

type B (PTPRB) inhibits brown adipocyte differentiation through regulation of VEGFR2

676

phosphorylation. J Microbiol Biotechnol. 2019.

682 683 684 685 686 687 688 689 690

oo

pr

e-

681

Jab1-mediated nuclear export of p53. BMB Rep. 2017;50:373-8. [39] Koivunen P, Lee S, Duncan CG, Lopez G, Lu G, Ramkissoon S, et al. Transformation by the (R)enantiomer of 2-hydroxyglutarate linked to EGLN activation. Nature. 2012;483:484-8.

Pr

680

[38] Lee EW, Oh W, Song HP, Kim WK. Phosphorylation of p53 at threonine 155 is required for

[40] Prensner JR, Chinnaiyan AM. Metabolism unhinged: IDH mutations in cancer. Nat Med. 2011;17:291-3.

[41] Ge K. Epigenetic regulation of adipogenesis by histone methylation. Biochim Biophys Acta.

al

679

Metabolic Reprogramming of Brown Adipose Tissue. Cell Rep. 2016;17:1008-21.

2012;1819:727-32.

[42] Mikkelsen TS, Xu Z, Zhang X, Wang L, Gimble JM, Lander ES, et al. Comparative epigenomic

rn

678

[37] Duteil D, Tosic M, Lausecker F, Nenseth HZ, Muller JM, Urban S, et al. Lsd1 Ablation Triggers

analysis of murine and human adipogenesis. Cell. 2010;143:156-69.

Jo u

677

f

674

[43] Seale P. Transcriptional Regulatory Circuits Controlling Brown Fat Development and Activation. Diabetes. 2015;64:2369-75.

691

[44] Yang Q, Liang X, Sun X, Zhang L, Fu X, Rogers CJ, et al. AMPK/alpha-Ketoglutarate Axis

692

Dynamically Mediates DNA Demethylation in the Prdm16 Promoter and Brown Adipogenesis.

693

Cell Metab. 2016;24:542-54.

694 695 696 697 698 699 700 701

[45] Kajimura S, Saito M. A new era in brown adipose tissue biology: molecular control of brown fat development and energy homeostasis. Annu Rev Physiol. 2014;76:225-49. [46] Mills EL, Pierce KA, Jedrychowski MP, Garrity R, Winther S, Vidoni S, et al. Accumulation of succinate controls activation of adipose tissue thermogenesis. Nature. 2018;560:102-6. [47] Yen KE, Bittinger MA, Su SM, Fantin VR. Cancer-associated IDH mutations: biomarker and therapeutic opportunities. Oncogene. 2010;29:6409-17. [48] Pan D, Fujimoto M, Lopes A, Wang YX. Twist-1 is a PPARdelta-inducible, negative-feedback regulator of PGC-1alpha in brown fat metabolism. Cell. 2009;137:73-86. 30

Journal Pre-proof 702 703 704 705

[49] Chao LC, Bensinger SJ, Villanueva CJ, Wroblewski K, Tontonoz P. Inhibition of adipocyte differentiation by Nur77, Nurr1, and Nor1. Mol Endocrinol. 2008;22:2596-608. [50] Kohli RM, Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature. 2013;502:472-9.

706

[51] Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, et al. Oncometabolite 2-hydroxyglutarate is a

707

competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011;19:17-

708

30.

709

[52] Zhuang L, Jang Y, Park YK, Lee JE, Jain S, Froimchuk E, et al. Depletion of Nsd2-mediated

710

histone H3K36 methylation impairs adipose tissue development and function. Nat Commun.

711

2018;9:1796. [53] Cho YW, Hong S, Jin Q, Wang L, Lee JE, Gavrilova O, et al. Histone methylation regulator PTIP

713

is required for PPARgamma and C/EBPalpha expression and adipogenesis. Cell Metab.

714

2009;10:27-39.

oo

f

712

[54] Lee J, Saha PK, Yang QH, Lee S, Park JY, Suh Y, et al. Targeted inactivation of MLL3 histone

716

H3-Lys-4 methyltransferase activity in the mouse reveals vital roles for MLL3 in adipogenesis.

717

Proc Natl Acad Sci U S A. 2008;105:19229-34.

720 721

e-

Pr

719

[55] Koh HJ, Lee SM, Son BG, Lee SH, Ryoo ZY, Chang KT, et al. Cytosolic NADP+-dependent isocitrate dehydrogenase plays a key role in lipid metabolism. J Biol Chem. 2004;279:39968-74. [56] Carey BW, Finley LW, Cross JR, Allis CD, Thompson CB. Intracellular alpha-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature. 2015;518:413-6.

al

718

pr

715

[57] TeSlaa T, Chaikovsky AC, Lipchina I, Escobar SL, Hochedlinger K, Huang J, et al. alpha-

723

Ketoglutarate Accelerates the Initial Differentiation of Primed Human Pluripotent Stem Cells.

724

Cell Metab. 2016;24:485-93.

726 727 728 729 730

Jo u

725

rn

722

[58] Monteiro MC, Sanyal M, Cleary ML, Sengenes C, Bouloumie A, Dani C, et al. PBX1: a novel stage-specific regulator of adipocyte development. Stem Cells. 2011;29:1837-48. [59] Caprio M, Feve B, Claes A, Viengchareun S, Lombes M, Zennaro MC. Pivotal role of the mineralocorticoid receptor in corticosteroid-induced adipogenesis. FASEB J. 2007;21:2185-94. [60] Lee H, Kang R, Bae S, Yoon Y. AICAR, an activator of AMPK, inhibits adipogenesis via the WNT/beta-catenin pathway in 3T3-L1 adipocytes. Int J Mol Med. 2011;28:65-71.

731

[61] Fernandez-Veledo S, Vazquez-Carballo A, Vila-Bedmar R, Ceperuelo-Mallafre V, Vendrell J.

732

Role of energy- and nutrient-sensing kinases AMP-activated protein kinase (AMPK) and

733

mammalian target of rapamycin (mTOR) in adipocyte differentiation. IUBMB Life.

734

2013;65:572-83.

735

[62] Vila-Bedmar R, Lorenzo M, Fernandez-Veledo S. Adenosine 5'-monophosphate-activated protein

736

kinase-mammalian target of rapamycin cross talk regulates brown adipocyte differentiation.

737

Endocrinology. 2010;151:980-92. 31

Journal Pre-proof 738

[63] Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts its anti-diabetic effects

739

through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J. 2000;348 Pt

740

3:607-14.

IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462:739-44.

f

748

[67] Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, et al. Cancer-associated

oo

747

transcriptional axis suppresses melanoma metastasis. Nature. 2016;537:422-6.

pr

746

[66] Luo C, Lim JH, Lee Y, Granter SR, Thomas A, Vazquez F, et al. A PGC1alpha-mediated

e-

745

2008;6:25-34.

Pr

744

[65] Blanquicett C, Roman J, Hart CM. Thiazolidinediones as anti-cancer agents. Cancer Ther.

al

743

2003;9:1-9.

rn

742

[64] Koeffler HP. Peroxisome proliferator-activated receptor gamma and cancers. Clin Cancer Res.

Jo u

741

32

Journal Pre-proof Figure Legends

750

Fig. 1. IDH1 is upregulated during brown fat differentiation and development. (A)

751

Protein levels of IDH1 and IDH2 during adipogenesis of immortalized brown preadipocytes

752

and primary brown preadipocytes. Numbers below the immunoblots indicate band intensity

753

(normalized to HSP90) quantified by using ImageJ software. (B) mRNA levels of Idh1 and

754

brown adipogenic genes during differentiation of immortalized brown preadipocytes (n = 3).

755

Data represent the mean ± SD. Statistical analysis was performed using one-way ANOVA. *p

756

< 0.05 and *p < 0.005 vs. brown preadipocytes (Day 0). (C) Time course of IDH1 and UCP1

757

protein expression during brown fat development from embryonic day 18 (E18), postnatal

758

days 1–6 (P1–6), and 12-week-old (12w) mice. IDH1 and UCP1 levels were measured by

759

western blot, with HSP90 used as a loading control. (D) mRNA levels of Idh1 and brown

760

adipogenic genes during various time course of brown fat development as determined by

761

qRT-PCR (n =2). Data represent the mean ± SD. Statistical analysis was performed using

762

one-way ANOVA. *p < 0.05 vs. E18. All experiments were repeated independently at least

763

three times and representative results are shown.

oo

pr

e-

Pr

al

rn Jo u

764

f

749

765

Fig. 2. Ectopic IDH1 expression inhibits differentiation of brown preadipocytes. (A) The

766

efficiency of retroviral transduction and monitoring of IDH1 levels by fluorescence

767

microscopy. Retrovirus-transduced cells were selected by GFP-positive cell sorting. (B)

768

IDH1 and UCP1 levels were measured by western blot, with HSP90 used as a loading control.

769

(C) Oil-Red-O staining of lipid droplets after differentiation of Idh1-overexpressing brown

770

adipocytes. (D) For quantitation of lipid content, Oil-red-O stain was eluted from cells with

771

isopropanol, and measurements were taken at 490 nm (n = 3). Data represent the mean ± SD.

772

(E) mRNA levels of brown adipogenic genes in mature brown adipocytes as determined by

773

qRT-PCR and normalized to TBP (n = 3). Data represent the mean ± SD. Statistical analysis 33

Journal Pre-proof 774

was performed using a two-tailed Student's t test. *p < 0.05, **p < 0.005, and ***p < 0.0005

775

vs. Vector. The data shown are representative results of at least three independent

776

experiments.

777

Fig. 3. IDH1 suppression inhibits brown adipocyte differentiation. (A) RFP levels were

779

monitored by fluorescence microscopy. Brown preadipocytes were transduced with retroviral

780

vectors expressing pSIREN-RetroQ-DsRed. (B) Efficiency of Idh1 knockdown in IDH1-

781

suppressed brown preadipocytes according to qRT-PCR (n = 2). Data represent the mean ±

782

SD. Statistical analysis was performed using one-way ANOVA. (C) IDH1 levels according to

783

western blot. Relative amount of IDH1 protein was calculated using ImageJ software and

784

normalized to HSP90. (D) Oil-Red-O staining of lipid droplets after differentiation of IDH1-

785

suppressed brown adipocytes and quantification of lipid accumulation (n = 3). Data represent

786

the mean ± SD. Statistical analysis was performed using a two-tailed Student's t test. (E)

787

IDH1 and UCP1 levels according to western blot. Numbers below the immunoblots indicate

788

band intensity (normalized to HSP90) quantified by using ImageJ software. n.d., not detected.

789

(F) mRNA levels of brown adipogenic genes according to qRT-PCR and normalized to TBP

790

(n = 3). Data represent the mean ± SD. Statistical analysis was performed using a two-tailed

791

Student's t test. *p < 0.05 and **p < 0.005 vs. SCR. The data shown are representative of

792

three independent experiments.

Jo u

rn

al

Pr

e-

pr

oo

f

778

793 794

Fig. 4. α-KG negatively regulates brown adipogenesis at the early stage of

795

differentiation. (A) α-KG levels following Idh1 overexpression or knockdown monitored

796

during brown adipocyte differentiation (n = 3). Data represent the mean ± SD. Statistical

797

analysis was performed using a two-tailed Student's t test. *p < 0.05, **p < 0.005, and ***p <

798

0.0005 vs. control (Vector or SCR). (B) Cell-permeable α-KG (dm-α-KG, 1 mM) was 34

Journal Pre-proof administered to brown preadipocytes, and α-KG levels were measured according to a time

800

course (n = 3). Data represent the mean ± SD. Statistical analysis was performed using one-

801

way ANOVA. n.s., not significant. (C) Oil-Red-O staining of lipid droplets after

802

differentiation of brown preadipocytes treated with dm-α-KG for 2 days (days 0–2, 2–4, and

803

4–6) and for 6 days (days 0–6) every 24 h. (D) mRNA levels of brown adipogenic genes in

804

dm-α-KG-treated mature brown adipocytes according to qRT-PCR and normalized to TBP (n

805

= 3). Statistical analysis was performed using one-way ANOVA. *p < 0.05, **p < 0.005, and

806

***p < 0.0005 vs. Vehicle. (E) Protein levels of UCP1, PGC1α, PRDM16, and PPARγ were

807

measured by western blot. Numbers below the immunoblots indicate band intensity

808

(normalized to HSP90) quantified by using ImageJ software. n.d., not detected. Similar

809

results were obtained in at least three independent experiments.

e-

pr

oo

f

799

Pr

810

Fig. 5. The effects of α-KG and Idh1 overexpression or knockdown on brown adipocyte

812

differentiation. Brown preadipocytes were treated with dm-α-KG (1 mM) during the early

813

stage of differentiation (days 0–2) or daily (days 0–6). (A, C) Oil-Red-O staining of lipid

814

droplets after differentiation of α-KG-treated Idh1-modulated brown adipocytes. (B, D)

815

mRNA levels of brown adipogenic genes according to qRT-PCR (n = 3). Data represent the

816

mean ± SD. Statistical analysis was performed using two-way ANOVA. *p < 0.05, **p <

817

0.005, and ***p < 0.0005 vs. control (Vehicle-treated control cells). ###p < 0.0005. All

818

experiments were repeated independently at least three times and representative results are

819

shown.

Jo u

rn

al

811

820 821

Fig. 6. α-KG controls epigenetic modification via histone methylation during brown

822

adipocyte differentiation. (A) Levels of H3K4me3, H3K9me3, H3K27me3, and H3K36me3

823

measured during brown adipocyte differentiation for 12 h, 24 h, and 48 h. Numbers below the 35

Journal Pre-proof immunoblots indicate band intensity (normalized to Histone H3) quantified by using ImageJ

825

software. (B) mRNA levels of brown adipogenic genes were monitored during brown

826

adipocyte differentiation in the presence of dm-α-KG (n = 3). Data represent the mean ± SD.

827

Statistical analysis was performed using two-way ANOVA. **p < 0.005 and ***p < 0.0005

828

vs. control (Vehicle-treated cells on day 0). #p < 0.05 and ##p < 0.005. n.s., not significant.

829

(C, D) Levels of histone H3 lysine methylation on the second day were examined during

830

differentiation of dm-α-KG-treated Idh1-overexpressing or -knockdown brown adipocytes. (E,

831

F) Enrichment of H3K4me3 and H3K36me3 levels in the promoters of adipogenic genes on

832

the second day of brown adipocyte differentiation in the presence of dm-α-KG (n = 3). Data

833

represent the mean ± SD. Statistical analysis was performed using two-way ANOVA. **p <

834

0.005 and ***p < 0.0005 vs. control (Vehicle-treated control cells). #p < 0.05 and ###p <

835

0.0005. The data shown are representative results of at least three independent experiments.

Pr

e-

pr

oo

f

824

836

Fig. 7. α-KG administration decreases adipogenic gene expression in brown adipose

838

tissues of mice. (A) 8-week-old male C57BL/6 mice were injected intraperitoneally with

839

saline and 0.5, 1, and 2 g/kg of dm-α-KG (n = 4). The mice were sacrificed 3 hours post

840

injections to dissect brown adipose tissues. α-KG levels were measured in brown adipose

841

tissues of mice injected with saline or α-KG. Data represent the mean ± SD. Statistical

842

analysis was performed using one-way ANOVA. *p < 0.05 vs. control (Saline-administered

843

mice) (B) mRNA levels of adipogenic genes in brown adipose tissues after 3 days of daily

844

intraperitoneal injection with 1 g/kg α-KG into C57BL/6 mice (n = 5). Data represent the

845

mean ± SD. Statistical analysis was performed using a two-tailed Student's t test. **p < 0.005

846

and ***p < 0.0005 vs. control (Saline-administered mice). n.s., not significant. (C) Protein

847

levels of UCP1, PGC1α, PRDM16, and PPARγ were monitored by western blot in brown

Jo u

rn

al

837

36

Journal Pre-proof 848

adipose tissues of mice injected with saline or α-KG for 3 days. (D) Levels of H3K4me3,

849

H3K9me3, H3K27me3, and H3K36me3 were measured in brown adipose tissues of saline or

850

α-KG administered mice. (E) Enrichment of H3K4me3 and H3K36me3 levels at the

851

promoters of brown adipocyte-specific genes in brown adipose tissues of mice injected with

852

saline or α-KG for 3 days. Data represent the mean ± SD. Statistical analysis was performed

853

using a two-tailed Student's t test. *p < 0.05 vs. control (Saline-administered mice). n.s., not

854

significant.

oo

f

855

Fig. 8. The AMPK activator metformin reduces brown adipogenesis. (A) Metformin (1

857

mM) was administered to brown preadipocytes, and AMPK and phosphorylated-AMPK

858

levels were measured by western blot. Relative amount of phosphorylated-AMPK was

859

quantified using ImageJ software and normalized to total AMPK. (B–D) The effects of

860

metformin on brown adipogenesis. Brown preadipocytes were treated with 1 mM metformin

861

every 48 h for 6 days. (B) α-KG levels were monitored in metformin-treated brown

862

adipocytes (n = 3). Data represent the mean ± SD. Statistical analysis was performed using

863

two-way ANOVA. **p < 0.005 and ***p < 0.0005 vs. control (Vehicle-treated cells on day 0).

864

##p < 0.005 and ###p < 0.0005. (C) Oil-Red-O staining of lipid droplets and quantification of

865

lipid accumulation in mature brown adipocytes in the presence or absence of metformin (n =

866

3). Data represent the mean ± SD. (D) mRNA levels of brown adipogenic genes in mature

867

adipocytes in the presence of metformin (n = 3). Data represent the mean ± SD. Statistical

868

analysis was performed using two-tailed Student's t test. *p < 0.05 vs. Vehicle. (E) Protein

869

levels in metformin-treated mature brown adipocytes. (F) Oil-Red-O staining of lipid droplets

870

and quantification of lipid accumulation in differentiated C3H10T1/2 MSCs after brown

871

adipogenesis (n = 3). Data represent the mean ± SD. Statistical analysis was performed using

872

a two-tailed Student's t test. n.s., not significant. (G) mRNA levels of brown adipogenic genes

Jo u

rn

al

Pr

e-

pr

856

37

Journal Pre-proof 873

during brown adipogenesis of C3H10T1/2 MSCs treated with or without 1mM dm-α-KG (n =

874

3). Data represent the mean ± SD. Statistical analysis was performed using a two-way

875

ANOVA. *p < 0.05, *p < 0.005, and ***p < 0.0005 vs. control (Vehicle-treated cells on day

876

0). #p < 0.05 and ##p < 0.005. The data shown are representative of three independent

877

experiments.

878

Fig. 9. IDH1 and α-KG in brown adipose tissues is regulated by cold-induced

880

thermogenesis. (A) mRNA levels of Idh1 and thermogenic genes in brown adipose tissues

881

from 11-week-old C57BL/6 mice housed at room temperature (RT) or exposed to cold (5°C)

882

for the indicated time points (n = 3). Data represent the mean ± SD. Statistical analysis was

883

performed using one-way ANOVA. *p < 0.05 and **p < 0.005 vs. control (RT). (B) Protein

884

levels of IDH1, UCP1, and PGC1α from brown adipose tissues of mice exposed to cold (5°C)

885

for the indicated durations. (C) α-KG levels were measured in brown adipose tissues of mice

886

upon cold exposure. Data represent the mean ± SD. Statistical analysis was performed using

887

one-way ANOVA. **p < 0.005 vs. control (RT). (D) Levels of histone methylation, such as

888

H3K4me3, H3K9me3, H3K27me3, and H3K36me3, were analyzed in in brown adipose

889

tissues of mice during cold exposure.

oo

pr

e-

Pr

al

rn

Jo u

890

f

879

38

Journal Pre-proof Author contributions

892

HSK, JHL, K-JO, EWL, SCL, K-HB, and WKK conceived the study design, and data

893

interpretation. HSK, JHL, K-JO, EWL, K-YP, and JMS performed experiments. HSK, JHL,

894

JMS, K-HB, and WKK contributed to writing the manuscript. K-JO, EWL, BSH, J-KM, S-

895

WC, and SCL critically reviewed the study and gave the necessary suggestions. All authors

896

read and approved the final version of the manuscript.

Jo u

rn

al

Pr

e-

pr

oo

f

891

39

Figure 1

Figure 2

Figure 3

Figure 4

Figure 5

Figure 6

Figure 7

Figure 8

Figure 9