Epigallocatechin Gallate Inhibits Hepatic Glucose Production in

Mar 15, 2019 - We further investigated the effects of EGCG on glucagon-stimulated cAMP/PKA signaling pathway. EGCG reduced p-PKA-T197/T-PKA and ...
0 downloads 0 Views 3MB Size
Subscriber access provided by UNIV OF CAMBRIDGE

Bioactive Constituents, Metabolites, and Functions

Epigallocatechin gallate inhibits hepatic glucose production in primary hepatocytes via downregulating PKA signaling pathway and transcriptional factor FoxO1 Xiaopeng Li, Yunmei Chen, Zheng Shen, Quan Pan, Wanbao Yang, Hui Yan, Weiqi Ai, Liao Wang, and Shaodong Guo J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.9b00395 • Publication Date (Web): 15 Mar 2019 Downloaded from http://pubs.acs.org on March 19, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 40

Journal of Agricultural and Food Chemistry

1

Epigallocatechin gallate inhibits hepatic glucose production in

2

primary hepatocytes via downregulating PKA signaling pathway and

3

transcriptional factor FoxO1

4

Xiaopeng Li

†,‡ ,

Yunmei Chen †# , James Zheng Shen † , Quan Pan † , Wanbao Yang † ,

5

Hui Yan†, Huimin Liu†, Weiqi Ai†, Wang Liao†, Shaodong Guo*,†

6

†Department of Nutrition and Food Science, College of Agriculture and Life Science,

7

Texas A&M University, College Station, TX 77843, USA

8

‡ College of Food Science and Technology, Huazhong Agricultural University,

9

Wuhan, 430070, China

10

# School

of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003,

11

China

12

*Corresponding

13

Shaodong Guo

14

Department of Nutrition and Food Science

15

College of Agriculture and Life Sciences, Texas A&M University

16

373 Olsen Blvd., Cater-Mattil Hall Rm. 123A, TAMU2253,

17

College Station, TX 77843, USA. Tel: 979-845-0850

18

Keywords

19

(-)-Epigallocatechin gallate / FoxO1 / Hepatic glucose production (HGP) / Glucagon /

20

AMPK

author:

Fax: 979-862-6842

21 22

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

23

ABSTRACT: Forkhead/winged helix transcription factor O-class member 1 (FoxO1)

24

is a key mediator of insulin and glucagon signaling in control of glucose homeostasis.

25

Although epigallocatechin gallate (EGCG) has attracted interests owing to its

26

potential to combat hyperglycemic diabetes, molecular mechanisms underlying its

27

antihyperglycemic effect, in particular the effect on FoxO1 is poorly understand. This

28

study aims to assess the impact of EGCG on the glucagon signaling pathway in

29

regulating glucose metabolism. Primary hepatocytes from wild-type (WT),

30

liver-specific FoxO1 knock out (FKO) and FoxO1-S273D knock-in (KI) mice were

31

isolated, cultured, and treated with EGCG or/and glucagon. Our data showed the

32

treatment of 10 μM EGCG for 6 h decreased hepatic glucose production by 20% and

33

23% in WT and FKO primary hepatocytes, respectively. EGCG repressed both

34

gluconeogenesis and glycogenolysis in primary hepatocytes, coupled with activating

35

AMPK. In addition, EGCG decreased mitochondrial oxygen consumption. We further

36

investigated the effects of EGCG on glucagon-stimulated cAMP/PKA signaling

37

pathway. EGCG reduced p-PKA-T197/T-PKA and p-CREB-S133/T-CREB level by

38

39% and 20%, blocked p-FoxO1-S273 and suppressed nuclear FoxO1 translocation,

39

suggesting that FoxO1 and CREB were possible downstream targets. A novel

40

mechanism of EGCG in restraining hepatic glucose production (HGP) is through

41

antagonizing glucagon signaling and suppressing FoxO1 via Ser273. EGCG may

42

serve as a promising compound for regulating glucose homeostasis.

43 44

ACS Paragon Plus Environment

Page 2 of 40

Page 3 of 40

Journal of Agricultural and Food Chemistry

45

■INTRODUCTION

46

Diabetes is characterized by the deregulation of glucose homeostasis partly owing to

47

increased hepatic glucose production (HGP). And now diabetes is becoming a global

48

health issue. An estimated 30.2 million people in the United States in 2017 according

49

to the International Diabetes Federation (IDF) and more than 86 million Americans

50

are considered pre-diabetic.1 In China, the number of people with diabetes reached

51

114.4 million in 2017. The healthcare expenditure of people with diabetes is expected

52

to continue to grow. The highest yearly cost per person with diabetes is the US with

53

11,638 USD (IDF, 2017, http://diabetesatlas.org). Moreover, diabetes dramatically

54

elevated comorbidities of other chronic health issues, including cardiovascular

55

disease, kidney disease, and diabetic eye disease.2

56

Two important transcriptional factors regulate gluconeogenesis: FoxO1 and element

binding

protein

(CREB).1,

3

57

cAMP-responsible

58

gluconeogenic genes expression, PEPCK, and G-6-Pase, as well as contributes to the

59

regulation of cell metabolism.4 As a transcriptional factor in response to insulin,

60

FoxO1 is inhibited by phosphorylating at S256 in human, equivalent to S253 of

61

FoxO1 in mice, through insulin/Akt signaling pathway.5 The phosphorylation of

62

FoxO1 at S256 resulted in promoting transport from nuclear to cytoplasmic and

63

reducing gluconeogenesis. In contrast, our lab has found that glucagon could improve

64

FoxO1 nuclear translocation and stability through phosphorylation of FoxO1-S273 by

65

PKA activation, suggesting that FoxO1-S273 also participated in glucagon/PKA

66

signaling pathway.6-7 CREB binding protein (CBP) complex is another transcriptional

ACS Paragon Plus Environment

FoxO1

promotes

Journal of Agricultural and Food Chemistry

67

factor regulated by cAMP/PKA through the activation at site Ser133 phosphorylation.

68

Such activation further leads to the recruitment of CREB-binding protein (CBP) to

69

activate the CREB-CBP complex in response to glucagon signaling. CREB-CBP

70

complex can directly bind to cAMP response element site (CRE) to induce PGC1α

71

transcription and its downstream target genes such as the rate-limiting

72

gluconeogenetic enzymes genes, PEPCK and G-6-Pase.8-9 Additionally, insulin can

73

directly abrogate the cAMP signaling pathway by phosphorylation CBP at Ser 436,

74

interfering the binding of CBP to CREB.10 Metformin has shown suppressed the

75

gluconeogenesis through phosphorylation of CBP at Ser 436 by AMPK activation.11

76

EGCG, a major polyphenol from green tea, has been investigated with the

77

prevention of insulin resistance and diabetes.12-16 The plasma concentration of EGCG

78

after tea catechin intake could reach to 10 μM in animal study (10 mg/kg intravenous

79

administration).17 After a high dose of tea catechins in human oral administration, the

80

EGCG concentration in plasma could reach around 10 μM.18 Previous studies

81

demonstrated EGCG suppressed HGP through activating AMPK in primary

82

hepatocytes.19-20 AMPK activation reduced glucagon-stimulated cAMP signaling via

83

activation of cyclic nucleotide phosphodiesterase 4B (PDE4B).21 AMPK activation

84

also suppresses hepatic gluconeogenesis by destroying CREB-CBP complex through

85

CBP-S436 phosphorylation and interfering FoxO1 by class Ⅱ a histone deacetylases

86

(HDACs) phosphorylation.11,

87

gluconeogenesis. Metformin also inhibited hepatic gluconeogenesis in mice lacking

88

hepatic AMPK through changing the energy state.23 Meanwhile, EGCG has also been

22

However, AMPK is not necessary for regulating

ACS Paragon Plus Environment

Page 4 of 40

Page 5 of 40

Journal of Agricultural and Food Chemistry

89

reported to inhibit ATP synthase and downregulate mitochondrial complex I-V

90

activities, resulting in accumulating of ADP or AMP.24 AMP could directly activate

91

AMPK and bind to the P-site at the adenylate cyclase and suppress its activity,

92

causing a decrease accumulation of cAMP once stimulated by glucagon.25-26 The

93

inhibition of mitochondrial function may directly or indirectly interfere cAMP/PKA

94

signaling pathway. Thus, mitochondria seem to be at the crossroad in regulating

95

AMPK and cAMP/PKA signaling.

96

Under fasting condition, glucagon is secreted and activates cAMP/PKA signaling,

97

resulting in elevated gene expression of glycogenolytic and gluconeogenetic enzymes.

98

7

99

elusive. Based on these backgrounds, our objective was to determine the underlying

100

purpose of EGCG on cAMP/PKA/FoxO1 signaling pathway in regulating glucose

101

metabolism.

However, the role of EGCG in glucose homeostasis related to cAMP/PKA remains

102 103

■MATERIALS AND METHODS

104

Chemicals and antibodies

105

EGCG, glucagon, and the CBP-CREB interaction inhibitor were obtained from

106

Millpore Sigma (MA, USA). Metformin was got from Enzo, and PKA inhibitor H89

107

was obtained from Tocris (MN, USA).

108

(MA, USA). p-AMPK-T172, AMPKα, p-CREB-S133, CREB, p-Akt-S473, Akt,

109

p-PKA-T197, PKA, T-FoxO1, Histone, and GAPDH were bought from Cell

110

Signaling Technology (MA, USA). p-FoxO1-S273 was generated in our lab as

Compound C was purchased from Abcam

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

111

previously reported 7. NE-PER Nuclear and cytoplasmic extraction reagents and

112

pierce lactate dehydrogenase (LDH) cytotoxicity assay kit were supplied by Thermo

113

Scientific (MA, USA).

114

Isolation of primary hepatocytes

115

Mice deficient for FoxO1 gene in the liver (FKO mice) and FoxO1-S273D KI mice

116

were generated as previously described.7, 27 All mice used for isolation of hepatocytes

117

were kept in a controlled environment (22 ℃ and 65 ± 3% humidity) and fed with

118

normal chow diet. Animal protocols were approved by institutional animal care and

119

use committee (IACUC) at Texas A&M University (2015-0316). Under anesthesia

120

state with isoflurane, livers were infused with Hanks' balanced solution containing 10

121

mM Hepes, 0.5 mM EGTA, 5.5 mM glucose and 1% penicillin-streptomycin

122

(perfusion solutionⅠ) at 5.5 ml/min for 5 min, followed by Hanks' balanced solution

123

containing 0.055% collagenase (Worthington, typeⅡ), 10 mM Hepes, 1.5 mM CaCl2,

124

5.5 mM glucose and 1% penicillin-streptomycin ( perfusion solutionⅡ) at 5.5 ml/min

125

for 5 min. Livers were collected and mashed slowly, and then hepatocytes were

126

purified with Percoll (Sigma-Aldrich, St. Louis, MO) and centrifugation. Hepatocytes

127

were plated onto collagen-coated 6 well plates at 3 × 105 cells/well in DMEM with

128

2% FBS for HGP study. The primary hepatocytes were kept at 37 ℃ with 5% CO2 in

129

the air.

130

Gluconeogenesis, glycogenolysis, and HGP study

131

For HGP studies, primary hepatocytes were cultured in DMEM with 2% FBS. After

132

3-4 hours attachment, cells were washed with warmed PBS for 3 times and the

ACS Paragon Plus Environment

Page 6 of 40

Page 7 of 40

Journal of Agricultural and Food Chemistry

133

medium was changed to HGP buffer (119 mM NaCl, 5.0 mM KCl, 2.6 mM KH2PO4,

134

2.6 mM MgSO4, 2.0 mM CaCl2, 24.6 mM NaHCO3, 10 mM HEPES, 0.5% BSA, 10

135

mM sodium L-lactate, and 5 mM pyruvate, pH=7.4). Subsequently, cells were treated

136

with EGCG, glucagon or both. It should be noted that the CREB-CBP interaction

137

inhibitor (40 μM) and compound C (5 μM) were used to pre-treat hepatocytes for 30

138

min before adding EGCG or glucagon in this study.

139

To test the effects of EGCG on glycogenolysis and gluconeogenesis, HGP buffer

140

with or without substrates (pyruvates and sodium L-lactate) were used in this study.

141

The glucose production hydrolyzed from glycogenolysis was determined in the

142

absence

143

gluconeogenesis is defined as the increase between the glucose production in HGP

144

buffer and HGP buffer without substrates. Cell culture medium was collected at 1, 3,

145

and 6 h after treatment for glucose content determination by using Amplex Red

146

Glucose Assay kit (Invitrogen, Carlsbad, CA). Glucose production was normalized by

147

protein concentration in cell lysates determined by BCA assay (Invitrogen, Carlsbad,

148

CA).

149

LDH release assay

150

The primary hepatocytes were seeded as 4,000 cells/well in 100 μL of medium in a

151

96-well tissue culture plate, followed by the incubation at 37 ℃ and 5% CO2

152

overnight. Add 10 μL of PBS to one group of cells (spontaneous LDH activity

153

controls), and nothing to another group of cells (maximum LDH activity controls).

154

Add 10 μL of PBS containing different concentration of EGCG (1-200 μL) to the

of

pyruvate/lactate

(substrates).

The

ACS Paragon Plus Environment

glucose

production

from

Journal of Agricultural and Food Chemistry

Page 8 of 40

155

cells. Incubate the plate in an incubator for 3 h, and add 10 μL of lysis buffer (10×)

156

and mix by gentle tapping. After 45 min, transfer 50 μL of each medium to a 96-well

157

plate and transfer 50 μL of the reaction mixture to each sample well. Incubate the

158

plate at room temperature for 30 min protected from light and add 50 μL of stop

159

solution. The absorbance was measured at 490 nm and 680 nm, and subtract the 680

160

nm absorbance value (background) from the 490 nm absorbance before calculation

161

of % Cytotoxicity.

162

% Cytotoxicity =

EGCG - treated LDH activity - Spontaneous LDH activity Maximum LDH activity - Spontaneous LDH activity

× 100

163

AMPK activation

164

For AMPK activation test, primary hepatocytes were cultured in completed medium

165

(DMEM with 10% FBS). After 3 hours of attachment, cells were washed with

166

warmed PBS for 3 times and the medium was changed to DMEM (fasting medium).

167

Then the cells were cultured with/without glucagon and co-treated with or without

168

EGCG for 1 h and were collected for Western blot analysis.

169

Mitochondria activity measurement

170

Mitochondrial function was measured by monitoring oxygen consumption rate (OCR)

171

using XF Cell Mito Stress Test Kit (Agilent technology). Isolated primary hepatocytes

172

were seeded in an XF96 cell culture microplate. The primary hepatocytes were

173

cultured in completed medium (DMEM with 10% FBS). After 3 hours attachment,

174

cells were washed with warmed PBS for 3 times and the medium was changed to

175

DMEM (fasting medium). The primary hepatocytes were treated with EGCG or

176

metformin. Seahorse assay was run in XF96 Extracellular Flux Analyzer (Agilent

ACS Paragon Plus Environment

Page 9 of 40

Journal of Agricultural and Food Chemistry

177

technology). During the OCR measurement, cells were exposed sequentially to

178

Oligomycin (0.5 μmol/L), Carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone

179

(FCCP, 1 μmol/L), and Rotenone (0.5 μmol/L). The OCR measurements were

180

recorded after every injection. To normalize the results, the protein of each well was

181

quantified using BCA protein assay kit (Thermo Scientific, MA, USA).

182

Nuclear and cytoplasmic protein extraction

183

Nuclear and cytoplasmic proteins from primary hepatocytes were extracted by

184

NE-PER Nuclear and cytoplasmic extraction kit according to manufactory's

185

instruction. Primary hepatocytes were seeded in completed medium and fasted for 2 h

186

with DMEM, and then treated with EGCG for 1 h. The cells were washed with PBS

187

twice and harvested with trypsin-EDTA. Then centrifuge at 500 × g for 5 min and

188

wash the cells with PBS. Add 300 μL ice-cold CERⅠ, vortex and incubate the tubes

189

on ice for 10 min. Ice-cold CERⅡwas then added, mixed and centrifuged for 5 min at

190

16,000 × g. The supernatant section (cytoplasmic extract) was immediately

191

transferred to a clean pre-chilled tube. The insoluble fraction was suspended in 150

192

μL of ice-cold NER and continued vortex for 15 seconds every 10 min (a total of 40

193

min), and then centrifuged at 16,000 × g for 10 min. The supernatant was quickly

194

transferred to a clean tube and store at -80 ℃ until use.

195

Western blot analysis

196

Protein was extracted from primary hepatocytes in RIPA lysis buffer (Sigma-Aldrich,

197

St. Louis, MO). The nuclear and cytoplasmic protein were extracted using NE-PER

198

nuclear and cytoplasmic extraction reagents according to instructions provided by the

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 10 of 40

199

manufacturer (Thermo Scientific, MA, US). And the protein concentrations were

200

determined by BCA protein assay kit (Thermo Scientific, MA, USA). Subsequently,

201

protein extracts were diluted in sample buffer and heated for 10 min in boiling water

202

bath. The protein samples were separated by SDS-polyacrylamide gel electrophoresis

203

(SDS-PAGE), and then electro-transferred to a PVDF membrane (Millipore, Bedford,

204

MA, USA). The membranes were then blocked with 5% BSA in TBST for 2 h at

205

room temperature in a shaker and incubated overnight at 4 ℃

206

antibodies in TBST (p-AMPK, 1:1000; AMPK, 1:1000; p-PKA-T197, 1:1000;

207

T-PKA, 1:1000; p-Akt-S473, 1:1000; T-Akt, 1:1000; p-CREB-S133, 1:1000;

208

T-CREB, 1:1000; p-FoxO1-S273, 1:800; GAPDH, 1:1000), followed by the

209

incubation of horseradish peroxidase (HRP)-conjugated secondary antibodies

210

(1:5000) for 2 h at room temperature in a shaker. Immunoreactive bands were

211

visualized by the enhanced chemiluminescent reagents (ECL). The intensity of bands

212

was measured using ChemiDoc imaging system (Bio-Rad LaboratoriesInc., Hercules,

213

CA).

214

Quantitative Real-Time PCR

215

Primary hepatocyte RNA was extracted with Trizol reagent (Invitrogen, Carlsbad,

216

CA),

217

(Bio-RadLaboratoriesInc., Hercules, CA). Gene expression was measured using the

218

SYBR Green Supermix according to manufacturer's protocol in Real-time PCR

219

system (Bio-RadLaboratoriesInc., Hercules, CA). The mRNA levels were normalized

220

to 18S using double-delta Ct method and presented as relative fold changes. Real-time

and

cDNA

was synthesized

using

iScript

ACS Paragon Plus Environment

cDNA

with primary

Synthesis

kit

Page 11 of 40

Journal of Agricultural and Food Chemistry

221

PCR primers were listed in Supplementary Table 1.

222

Statistical analysis

223

All results are calculated as the mean ± SD. Statistical significance between two

224

groups was calculated using unpaired, two-tailed t-tests. Data form more than two

225

groups was compared using a one-way analysis of variance (ANOVA). Values of

226

p