1
|
Guo X, Li H, Zhu B, Wang X, Xu Q, Aquino E, Koo M, Li Q, Cai J, Glaser S, Wu C. HFD feeding for seven months abolishes STING disruption-driven but not female sex-based protection against hepatic steatosis and inflammation in mice. J Nutr Biochem 2025; 135:109770. [PMID: 39284534 PMCID: PMC11620956 DOI: 10.1016/j.jnutbio.2024.109770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024]
Abstract
Stimulator of interferon genes (STING) is positively correlated with the degrees of liver inflammation in human metabolic dysfunction-associated steatotic liver disease (MASLD). In addition, STING disruption alleviates MASLD in mice fed a high-fat diet (HFD) for 3 months (3-m-HFD). Here we investigated the role of the duration of dietary feeding in regulating MASLD in mice and explored the involvement of STING in sex differences in MASLD. Both male and female STING-disrupted (STINGgt) and wild-type C57BL/6J mice were fed an HFD for 3 or 7 months (7-m-HFD). Additionally, female STINGgt mice upon ovariectomy (OVX) and 3-m-HFD were analyzed for MASLD. Upon 3-m-HFD, STINGgt mice exhibited decreased severity of MASLD compared to control. However, upon 7-m-HFD, STINGgt mice were comparable with wild-type mice in body weight, fat mass, and MASLD. Regarding regulating the liver RNA transcriptome, 7-m-HFD increased the expression of genes indicating proinflammatory activation of various liver cells. Interestingly, the severity of MASLD in female mice was much lighter than in male mice, regardless of STING disruption. Upon OVX, female STINGgt mice showed significantly increased severity of MASLD relative to sham control but were comparable with male STINGgt mice. Upon treatment with 17-beta estradiol (E2), hepatocytes revealed decreased fat deposition while macrophages displayed decreases in lipopolysaccharide-induced phosphorylation of Nfkb p65 and Jnk p46 independent of STING. These results suggest that 7-m-HFD, without altering female sex-based protection, abolishes STING disruption-driven protection of MASLD, likely through causing proinflammatory activation of multiple types of liver cells to offset the effect of STING disruption.
Collapse
Affiliation(s)
- Xinlei Guo
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Xiaoxiao Wang
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Qian Xu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Eduardo Aquino
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Minji Koo
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - James Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Byran, Texas, USA.
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
2
|
Griesel BA, Olson AL. PFKFB3 protein in adipose tissue contributes to whole body glucose homeostasis. FASEB J 2024; 38:e70254. [PMID: 39659238 DOI: 10.1096/fj.202402070r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Age-dependent changes in adipose tissue are thought to play a role in development of insulin resistance. A major age-dependent change in adipose tissue is the downregulation of key proteins involved in carbohydrate metabolism. In the current study, we investigate the role of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) a key governor of the rate of glycolysis in adipocytes via the synthesis of fructose-2,6-bisphosphate that was significantly downregulated in aged mice. We employed an adipocyte-specific PFKFB3 mouse line to investigate the role of PFKFB3 on adipocyte function. In both aged mice and PFKFB3-knockout mice, we observed an increase in O-glcNAcylated proteins consistent with a shift in glucose metabolism toward the hexosamine biosynthetic pathway. Under chow-fed conditions, PFKFB3 knockout resulted in significantly smaller adipocyte area, but no difference in total fat mass. While glucose tolerance was unchanged under chow conditions, when mice were challenged with a 4 weeks high-fat feeding, PFKFB3 deletion led to a greater decrease in glucose tolerance as well as a significant increase in macrophage infiltration. These results indicate that perturbation of the glycolytic pathway in adipose tissue has multiple effects of adipocyte biology and may play a significant role in metabolic changes associated with aging. Results of this student support the notion that changes in glucose metabolism in adipose tissue impact whole-body metabolism.
Collapse
Affiliation(s)
- Beth A Griesel
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Ann Louise Olson
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| |
Collapse
|
3
|
Lee MJ, Kim J. The pathophysiology of visceral adipose tissues in cardiometabolic diseases. Biochem Pharmacol 2024; 222:116116. [PMID: 38460909 PMCID: PMC11407912 DOI: 10.1016/j.bcp.2024.116116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/21/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Central pattern of fat distribution, especially fat accumulation within the intraabdominal cavity increases risks for cardiometabolic diseases. Portal hypothesis combined with a pathological remodeling in visceral fat is considered the major etiological factor explaining the independent contribution of visceral obesity to cardiometabolic diseases. Excessive remodeling in visceral fat during development of obesity leads to dysfunctions in the depot, characterized by hypertrophy and death of adipocytes, hypoxia, inflammation, and fibrosis. Dysfunctional visceral fat secretes elevated levels of fatty acids, glycerol, and proinflammatory and profibrotic cytokines into the portal vein directly impacting the liver, the central regulator of systemic metabolism. These metabolic and endocrine products induce ectopic fat accumulation, insulin resistance, inflammation, and fibrosis in the liver, which in turn causes or exacerbates systemic metabolic derangements. Elucidation of underlying mechanisms that lead to the pathological remodeling and higher degree of dysfunctions in visceral adipose tissue is therefore, critical for the development of therapeutics to prevent deleterious sequelae in obesity. We review depot differences in metabolic and endocrine properties and expendabilities as well as underlying mechanisms that contribute to the pathophysiological aspects of visceral adiposity in cardiometabolic diseases. We also discuss impacts of different weight loss interventions on visceral adiposity and cardiometabolic diseases.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Hawaii 96822, USA.
| | - Jeehoon Kim
- Department of Sociology, Social Work, and Criminology, Idaho State University, Idaho 83209, USA
| |
Collapse
|
4
|
Joo HJ, D'Alessandro M, Oh G, Han S, Kim WJ, Chung GE, Jang Y, Lee JB, Lee C, Yang Y. Novel targets of β-TrCP cooperatively accelerate carbohydrate and fatty acid consumption. J Cell Physiol 2024; 239:e31095. [PMID: 37584358 DOI: 10.1002/jcp.31095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/29/2023] [Accepted: 07/25/2023] [Indexed: 08/17/2023]
Abstract
Cellular energy is primarily produced from glucose and fat through glycolysis and fatty acid oxidation (FAO) followed by the tricarboxylic acid cycle in mitochondria; energy homeostasis is carefully maintained via numerous feedback pathways. In this report, we uncovered a new master regulator of carbohydrate and lipid metabolism. When ubiquitin E3 ligase β-TrCP2 was inducibly knocked out in β-TrCP1 knockout adult mice, the resulting double knockout mice (DKO) lost fat mass rapidly. Biochemical analyses of the tissues and cells from β-TrCP2 KO and DKO mice revealed that glycolysis, FAO, and lipolysis were dramatically upregulated. The absence of β-TrCP2 increased the protein stability of metabolic rate-limiting enzymes including 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB3), adipose triglyceride lipase (ATGL), carnitine palmitoyltransferase 1A (CPT1A), and carnitine/acylcarnitine translocase (CACT). Our data suggest that β-TrCP is a potential regulator for total energy homeostasis by simultaneously controlling glucose and fatty acid metabolism and that targeting β-TrCP could be an effective strategy to treat obesity and other metabolic disorders.
Collapse
Affiliation(s)
- Hyun Jeong Joo
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, USA
| | - Matthew D'Alessandro
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, USA
| | - Gaeun Oh
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Sora Han
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Woo Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Ga Eun Chung
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Youjeong Jang
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Jung Bok Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Choogon Lee
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida, USA
| | - Young Yang
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| |
Collapse
|
5
|
Hu AJ, Li W, Dinh C, Zhang Y, Hu JK, Daniele SG, Hou X, Yang Z, Asara JM, Hu GF, Farmer SR, Hu MG. CDK6 inhibits de novo lipogenesis in white adipose tissues but not in the liver. Nat Commun 2024; 15:1091. [PMID: 38316780 PMCID: PMC10844593 DOI: 10.1038/s41467-024-45294-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/19/2024] [Indexed: 02/07/2024] Open
Abstract
Increased de novo lipogenesis (DNL) in white adipose tissue is associated with insulin sensitivity. Under both Normal-Chow-Diet and High-Fat-Diet, mice expressing a kinase inactive Cyclin-dependent kinase 6 (Cdk6) allele (K43M) display an increase in DNL in visceral white adipose tissues (VAT) as compared to wild type mice (WT), accompanied by markedly increased lipogenic transcriptional factor Carbohydrate-responsive element-binding proteins (CHREBP) and lipogenic enzymes in VAT but not in the liver. Treatment of WT mice under HFD with a CDK6 inhibitor recapitulates the phenotypes observed in K43M mice. Mechanistically, CDK6 phosphorylates AMP-activated protein kinase, leading to phosphorylation and inactivation of acetyl-CoA carboxylase, a key enzyme in DNL. CDK6 also phosphorylates CHREBP thus preventing its entry into the nucleus. Ablation of runt related transcription factor 1 in K43M mature adipocytes reverses most of the phenotypes observed in K43M mice. These results demonstrate a role of CDK6 in DNL and a strategy to alleviate metabolic syndromes.
Collapse
Affiliation(s)
- Alexander J Hu
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Wei Li
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Calvin Dinh
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Yongzhao Zhang
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Jamie K Hu
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- University of Miami Miller School of Medicine, Dermatology. 1295 NW 14th St. University of Miami Hospital South Bldg. Suites K-M, Miami, FL, USA
| | - Stefano G Daniele
- Yale School of Medicine, MD-PhD program, 333 Cedar St, New Haven, CT, USA
| | - Xiaoli Hou
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- Zhejiang Chinese Medical University, Center for Analysis and Testing, 548 Bin-Wen Road, Hangzhou, PR China
| | - Zixuan Yang
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
- TUFTS University Friedman School of Nutrition Science and Policy, TUFTS University, 150 Harrison Avenue, MA, Boston, USA
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Guo-Fu Hu
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Stephen R Farmer
- Boston University School of Medicine, Department of Biochemistry, 72E Concord St, Boston, MA, USA
| | - Miaofen G Hu
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA.
| |
Collapse
|
6
|
Talukdar FR, Escobar Marcillo DI, Laskar RS, Novoloaca A, Cuenin C, Sbraccia P, Nisticò L, Guglielmi V, Gheit T, Tommasino M, Dogliotti E, Fortini P, Herceg Z. Bariatric surgery-induced weight loss and associated genome-wide DNA-methylation alterations in obese individuals. Clin Epigenetics 2022; 14:176. [PMID: 36528638 PMCID: PMC9759858 DOI: 10.1186/s13148-022-01401-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Obesity is a multifactorial and chronic condition of growing universal concern. It has recently been reported that bariatric surgery is a more successful treatment for severe obesity than other noninvasive interventions, resulting in rapid significant weight loss and associated chronic disease remission. The identification of distinct epigenetic patterns in patients who are obese or have metabolic imbalances has suggested a potential role for epigenetic alterations in causal or mediating pathways in the development of obesity-related pathologies. Specific changes in the epigenome (DNA methylome), associated with metabolic disorders, can be detected in the blood. We investigated whether such epigenetic changes are reversible after weight loss using genome-wide DNA methylome analysis of blood samples from individuals with severe obesity (mean BMI ~ 45) undergoing bariatric surgery. RESULTS Our analysis revealed 41 significant (Bonferroni p < 0.05) and 1169 (false discovery rate p < 0.05) suggestive differentially methylated positions (DMPs) associated with weight loss due to bariatric surgery. Among the 41 significant DMPs, 5 CpGs were replicated in an independent cohort of BMI-discordant monozygotic twins (the heavier twin underwent diet-induced weight loss). The effect sizes of these 5 CpGs were consistent across discovery and replication sets (p < 0.05). We also identified 192 differentially methylated regions (DMRs) among which SMAD6 and PFKFB3 genes were the top hypermethylated and hypomethylated regions, respectively. Pathway enrichment analysis of the DMR-associated genes showed that functional pathways related to immune function and type 1 diabetes were significant. Weight loss due to bariatric surgery also significantly decelerated epigenetic age 12 months after the intervention (mean = - 4.29; p = 0.02). CONCLUSIONS We identified weight loss-associated DNA-methylation alterations targeting immune and inflammatory gene pathways in blood samples from bariatric-surgery patients. The top hits were replicated in samples from an independent cohort of BMI-discordant monozygotic twins following a hypocaloric diet. Energy restriction and bariatric surgery thus share CpGs that may represent early indicators of response to the metabolic effects of weight loss. The analysis of bariatric surgery-associated DMRs suggests that epigenetic regulation of genes involved in endothelial and adipose tissue function is key in the pathophysiology of obesity.
Collapse
Affiliation(s)
- Fazlur Rahman Talukdar
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - David Israel Escobar Marcillo
- Section of Mechanisms, Biomarkers and Models, Dept Environment and Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
- Obesity Center-Internal Medicine Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ruhina Shirin Laskar
- Nutrition and Metabolism Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Alexei Novoloaca
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Cyrille Cuenin
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | - Paolo Sbraccia
- Obesity Center-Internal Medicine Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Lorenza Nisticò
- Centre for Behavioral Sciences and Mental Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
| | - Valeria Guglielmi
- Obesity Center-Internal Medicine Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tarik Gheit
- Early Detection, Prevention, and Infections Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| | | | - Eugenia Dogliotti
- Section of Mechanisms, Biomarkers and Models, Dept Environment and Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
| | - Paola Fortini
- Section of Mechanisms, Biomarkers and Models, Dept Environment and Health, Istituto Superiore Di Sanità, Viale Regina Elena, No. 299, 00161 Rome, Italy
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research On Cancer (IARC), 150 Cours Albert Thomas, Lyon, France
| |
Collapse
|
7
|
Cheng C, Liu XH, He J, Gao J, Zhou JT, Fan JN, Jin X, Zhang J, Chang L, Xiong Z, Yu J, Li S, Li X. Apolipoprotein A4 Restricts Diet-induced Hepatic Steatosis via SREBF1-mediated Lipogenesis and Enhances IRS-PI3K-Akt Signaling. Mol Nutr Food Res 2022; 66:e2101034. [PMID: 35909347 DOI: 10.1002/mnfr.202101034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 07/03/2022] [Indexed: 11/12/2022]
Abstract
SCOPE Hepatic steatosis and insulin resistance (IR) are risk factors for many metabolic syndromes such as NAFLD and T2DM. ApoA4 improves glucose hemostasis by increasing glucose-stimulated insulin secretion and glucose uptake via PI3K-Akt activation in adipocytes. However, whether ApoA4 has an effect on hepatic steatosis or IR remains unclear. METHODS AND RESULTS ApoA4-knockout (KO) aggravates diet-induced obesity, hepatic steatosis and IR in mice promoted by increased hepatic lipogenesis gene expression based on RNA-seq data. Conversely, liver-specific overexpression of ApoA4 via AAV-ApoA4 transduction reverses the effect in ApoA4-KO mice, accompanied by suppressed hepatic lipogenesis, increased lipolysis, and fatty acid oxidation. Short-term treatment with recombinant ApoA4 protein improves glucose clearance and liver insulin sensitivity, and reduces hepatic lipogenesis gene expression in the absence of insulin. Moreover, in primary hepatocytes and a hepatic cell line, ApoA4 improves hepatic glucose uptake via IRS-PI3K-Akt signaling and decreases fat deposition and hepatic lipogenesis gene expression by inhibiting SREBF1 activity. CONCLUSION ApoA4 restricts hepatic steatosis by inhibiting SREBF1-mediated lipogenesis and improves insulin sensitivity and glucose uptake via IRS-PI3K-Akt signaling in the liver. These findings indicate that ApoA4 may serve as a therapeutic target for obesity-associated NAFLD. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Cheng Cheng
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Xiao-Huan Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, Institute of Digestive Diseases. The Second Affiliated Hospital, Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jing He
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jing Gao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jin-Ting Zhou
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jing-Na Fan
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Xi Jin
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jianbo Zhang
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Liao Chang
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Zijun Xiong
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Jun Yu
- OneHealth Technology Company, Xi'an, 710000, China
| | - Shengbin Li
- Bio-evidence Sciences Academy (BSA), Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China.,Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, Institute of Digestive Diseases. The Second Affiliated Hospital, Xi'an Jiaotong University, Western China Science & Technology Innovation Harbour, Xi'an, 710100, China
| |
Collapse
|
8
|
Bioinformatics study of the potential therapeutic effects of ginsenoside Rf in reversing nonalcoholic fatty liver disease. Biomed Pharmacother 2022; 149:112879. [PMID: 35358801 DOI: 10.1016/j.biopha.2022.112879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/12/2022] [Accepted: 03/23/2022] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Ginsenoside Rf, a tetracyclic triterpenoid only present in Panax ginseng, has been proven to relieve lipid metabolism and inflammatory reactions, which can be a potential treatment for nonalcoholic fatty liver disease (NAFLD). Therefore, this study aimed to reveal the underlying mechanisms of ginsenoside Rf in the treatment of early-stage NAFLD (NAFL) by using a bioinformatics method and biological experiments. METHODS Target genes associated with NAFL were screened from the Gene Expression Omnibus (GEO) database, a database repository of high-throughput gene expression data and hybridization arrays, chips, and microarrays. Subsequently, gene set enrichment analysis was performed by using Gene Ontology enrichment analysis tool. Then, the binding capacity between ginsenoside Rf and NAFL-related targets was evaluated by molecular docking. Finally, the FFA-induced HepG2 cell model treated with ginsenoside Rf was adopted to verify the effect of ginsenoside Rf and the related mechanisms. RESULTS There were 41 common differentially expressed genes in the GEO dataset. Gene Ontology and Reactome pathway enrichment analysis of the differentially expressed genes showed that many pathways could be related to the pathogenesis of NAFL, including those participating in the cytokine-mediated signaling pathway, G protein-coupled receptor signaling pathway, and response to lipopolysaccharide. Finally, the qRT-PCR analysis results indicated that ginsenoside Rf therapy could ameliorate the transcription of ANXA2, BAZ1A, DNMT3L and MMP9. CONCLUSION Our research discovered the relevant mechanisms and basic pharmacological effects of ginsenoside Rf in the treatment of NAFL. These results might facilitate the development of ginsenoside Rf as an alternative medication for NAFL.
Collapse
|
9
|
Liu T, Wang Y, Zhao M, Jiang J, Li T, Zhang M. Differential expression of aerobic oxidative metabolism-related proteins in diabetic urinary exosomes. Front Endocrinol (Lausanne) 2022; 13:992827. [PMID: 36187097 PMCID: PMC9515495 DOI: 10.3389/fendo.2022.992827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND As a metabolic disease, any abnormality in the aerobic oxidation pathway of glucose may lead to the occurrence of diabetes. This study aimed to investigate the changes in proteins related to aerobic oxidative metabolism in urinary exosomes of diabetic patients and normal controls of different ages, and to further verify their correlation with the pathogenesis of diabetes. METHODS Samples were collected, and proteomic information of urinary exosomes was collected by LC-MS/MS. ELISA was used to further detect the expression of aerobic and oxidative metabolism-related proteins in urinary exosomes of diabetic patients and normal controls of different ages, and to draw receiver operating characteristic (ROC) curve to evaluate its value in diabetes monitoring. RESULTS A total of 17 proteins involved in aerobic oxidative metabolism of glucose were identified in urinary exosome proteins. Compared with normal control, the expressions of PFKM, GAPDH, ACO2 and MDH2 in diabetic patients were decreased, and the expression of IDH3G was increased. The concentrations of PFKM, GAPDH and ACO2 in urinary exosomes were linearly correlated with the expression of MDH2 (P<0.05). These four proteins vary with age, with the maximum concentration in the 45-59 age group. PFKM, GAPDH, ACO2, and MDH2 in urinary exosomes have certain monitoring value. When used in combination, the AUC was 0.840 (95% CI 0.764-0.915). CONCLUSIONS In diabetic patients, aerobic oxidative metabolism is reduced, and the expression of aerobic oxidative metabolism-related proteins PFKM, GAPDH, ACO2, and MDH2 in urinary exosomes is reduced, which may become potential biomarkers for monitoring changes in diabetes.
Collapse
Affiliation(s)
- Tianci Liu
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Yizhao Wang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Man Zhao
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Jun Jiang
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
- Clinical Laboratory Medicine, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Tao Li
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Man Zhang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
- Clinical Laboratory Medicine, Peking University Ninth School of Clinical Medicine, Beijing, China
- *Correspondence: Man Zhang,
| |
Collapse
|
10
|
Yin W, Luo S, Xiao Z, Zhang Z, Liu B, Zhou Z. Latent autoimmune diabetes in adults: a focus on β-cell protection and therapy. Front Endocrinol (Lausanne) 2022; 13:959011. [PMID: 35992113 PMCID: PMC9389314 DOI: 10.3389/fendo.2022.959011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Latent autoimmune diabetes in adults (LADA) is a heterogeneous disease sharing some phenotypic, genetic, and immunological features with both type 1 and 2 diabetes. Patients with LADA have a relatively slow autoimmune process and more residual islet β-cell function at onset, allowing a time window to protect residual islet β cells and delay or inhibit disease progression. It is crucial to discover various heterogeneous factors affecting islet β-cell function for precise LADA therapy. In this review, we first describe the natural history of LADA. Thereafter, we summarize β-cell function-related heterogeneous factors in LADA, including the age of onset, body mass index, genetic background, and immune, lifestyle, and environmental factors. In parallel, we evaluate the impact of current hypoglycemic agents and immune intervention therapies for islet β-cell protection. Finally, we discuss the opportunities and challenges of LADA treatment from the perspective of islet β-cell function protection.
Collapse
|
11
|
Recent Advances in Adipose Tissue Dysfunction and Its Role in the Pathogenesis of Non-Alcoholic Fatty Liver Disease. Cells 2021; 10:cells10123300. [PMID: 34943809 PMCID: PMC8699427 DOI: 10.3390/cells10123300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a serious ongoing health problem that significantly increases the incidence of nonalcoholic fatty liver disease (NAFLD). During obesity, adipose tissue dysfunction is obvious and characterized by increased fat deposition (adiposity) and chronic low-grade inflammation. The latter has been implicated to critically promote the development and progression of NAFLD, whose advanced form non-alcoholic steatohepatitis (NASH) is considered one of the most common causes of terminal liver diseases. This review summarizes the current knowledge on obesity-related adipose dysfunction and its roles in the pathogenesis of hepatic steatosis and inflammation, as well as liver fibrosis. A better understanding of the crosstalk between adipose tissue and liver under obesity is essential for the development of new and improved preventive and/or therapeutic approaches for managing NAFLD.
Collapse
|
12
|
Guo X, Zheng J, Zhang S, Jiang X, Chen T, Yu J, Wang S, Ma X, Wu C. Advances in Unhealthy Nutrition and Circadian Dysregulation in Pathophysiology of NAFLD. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2021; 2:691828. [PMID: 36994336 PMCID: PMC10012147 DOI: 10.3389/fcdhc.2021.691828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022]
Abstract
Unhealthy diets and lifestyle result in various metabolic conditions including metabolic syndrome and non-alcoholic fatty liver disease (NAFLD). Much evidence indicates that disruption of circadian rhythms contributes to the development and progression of excessive hepatic fat deposition and inflammation, as well as liver fibrosis, a key characteristic of non-steatohepatitis (NASH) or the advanced form of NAFLD. In this review, we emphasize the importance of nutrition as a critical factor in the regulation of circadian clock in the liver. We also focus on the roles of the rhythms of nutrient intake and the composition of diets in the regulation of circadian clocks in the context of controlling hepatic glucose and fat metabolism. We then summarize the effects of unhealthy nutrition and circadian dysregulation on the development of hepatic steatosis and inflammation. A better understanding of how the interplay among nutrition, circadian rhythms, and dysregulated metabolism result in hepatic steatosis and inflammation can help develop improved preventive and/or therapeutic strategies for managing NAFLD.
Collapse
Affiliation(s)
- Xin Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Xin Guo, ; Chaodong Wu,
| | - Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Shixiu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofan Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ting Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Jiayu Yu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Shu'e Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaomin Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- *Correspondence: Xin Guo, ; Chaodong Wu,
| |
Collapse
|
13
|
Griesel BA, Matsuzaki S, Batushansky A, Griffin TM, Humphries KM, Olson AL. PFKFB3-dependent glucose metabolism regulates 3T3-L1 adipocyte development. FASEB J 2021; 35:e21728. [PMID: 34110658 PMCID: PMC8205188 DOI: 10.1096/fj.202100381rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/08/2021] [Accepted: 05/25/2021] [Indexed: 11/11/2022]
Abstract
Proliferation and differentiation of preadipocytes, and other cell types, is accompanied by an increase in glucose uptake. Previous work showed that a pulse of high glucose was required during the first 3 days of differentiation in vitro, but was not required after that. The specific glucose metabolism pathways required for adipocyte differentiation are unknown. Herein, we used 3T3-L1 adipocytes as a model system to study glucose metabolism and expansion of the adipocyte metabolome during the first 3 days of differentiation. Our primary outcome measures were GLUT4 and adiponectin, key proteins associated with healthy adipocytes. Using complete media with 0 or 5 mM glucose, we distinguished between developmental features that were dependent on the differentiation cocktail of dexamethasone, insulin, and isobutylmethylxanthine alone or the cocktail plus glucose. Cocktail alone was sufficient to activate the capacity for 2-deoxglucose uptake and glycolysis, but was unable to support the expression of GLUT4 and adiponectin in mature adipocytes. In contrast, 5 mM glucose in the media promoted a transient increase in glucose uptake and glycolysis as well as a significant expansion of the adipocyte metabolome and proteome. Using genetic and pharmacologic approaches, we found that the positive effects of 5 mM glucose on adipocyte differentiation were specifically due to increased expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a key regulator of glycolysis and the ancillary glucose metabolic pathways. Our data reveal a critical role for PFKFB3 activity in regulating the cellular metabolic remodeling required for adipocyte differentiation and maturation.
Collapse
Affiliation(s)
- Beth A Griesel
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | | | - Timothy M Griffin
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kenneth M Humphries
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ann Louise Olson
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
14
|
Xu H, Zhu B, Li H, Jiang B, Wang Y, Yin Q, Cai J, Glaser S, Francis H, Alpini G, Wu C. Adipocyte inducible 6-phosphofructo-2-kinase suppresses adipose tissue inflammation and promotes macrophage anti-inflammatory activation. J Nutr Biochem 2021; 95:108764. [PMID: 33964465 DOI: 10.1016/j.jnutbio.2021.108764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/11/2021] [Accepted: 04/16/2021] [Indexed: 01/22/2023]
Abstract
Obesity-associated inflammation in white adipose tissue (WAT) is a causal factor of systemic insulin resistance. To better understand how adipocytes regulate WAT inflammation, the present study generated chimeric mice in which inducible 6-phosphofructo-2-kinase was low, normal, or high in WAT while the expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (Pfkfb3) was normal in hematopoietic cells, and analyzed changes in high-fat diet (HFD)-induced WAT inflammation and systemic insulin resistance in the mice. Indicated by proinflammatory signaling and cytokine expression, the severity of HFD-induced WAT inflammation in WT → Pfkfb3+/- mice, whose Pfkfb3 was disrupted in WAT adipocytes but not hematopoietic cells, was comparable with that in WT → WT mice, whose Pfkfb3 was normal in all cells. In contrast, the severity of HFD-induced WAT inflammation in WT → Adi-Tg mice, whose Pfkfb3 was over-expressed in WAT adipocytes but not hematopoietic cells, remained much lower than that in WT → WT mice. Additionally, HFD-induced insulin resistance was correlated with the status of WAT inflammation and comparable between WT → Pfkfb3+/- mice and WT → WT mice, but was significantly lower in WT → Adi-Tg mice than in WT → WT mice. In vitro, palmitoleate decreased macrophage phosphorylation states of Jnk p46 and Nfkb p65 and potentiated the effect of interleukin 4 on suppressing macrophage proinflammatory activation. Taken together, these results suggest that the Pfkfb3 in adipocytes functions to suppress WAT inflammation. Moreover, the role played by adipocyte Pfkfb3 is attributable to, at least in part, palmitoleate promotion of macrophage anti-inflammatory activation.
Collapse
Affiliation(s)
- Hang Xu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA; Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Boxiong Jiang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yina Wang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiongli Yin
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - James Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Shannon Glaser
- Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas, USA
| | - Heather Francis
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, Indiana, USA; Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, Indiana, USA; Richard L. Roudebush VA Medical Center, Indianapolis, Indiana, USA
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
15
|
Zhu B, Guo X, Xu H, Jiang B, Li H, Wang Y, Yin Q, Zhou T, Cai JJ, Glaser S, Meng F, Francis H, Alpini G, Wu C. Adipose tissue inflammation and systemic insulin resistance in mice with diet-induced obesity is possibly associated with disruption of PFKFB3 in hematopoietic cells. J Transl Med 2021; 101:328-340. [PMID: 33462362 PMCID: PMC7897240 DOI: 10.1038/s41374-020-00523-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/13/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity-associated inflammation in white adipose tissue (WAT) is a causal factor of systemic insulin resistance; however, precisely how immune cells regulate WAT inflammation in relation to systemic insulin resistance remains to be elucidated. The present study examined a role for 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) in hematopoietic cells in regulating WAT inflammation and systemic insulin sensitivity. Male C57BL/6J mice were fed a high-fat diet (HFD) or low-fat diet (LFD) for 12 weeks and examined for WAT inducible 6-phosphofructo-2-kinase (iPFK2) content, while additional HFD-fed mice were treated with rosiglitazone and examined for PFKFB3 mRNAs in WAT stromal vascular cells (SVC). Also, chimeric mice in which PFKFB3 was disrupted only in hematopoietic cells and control chimeric mice were also fed an HFD and examined for HFD-induced WAT inflammation and systemic insulin resistance. In vitro, adipocytes were co-cultured with bone marrow-derived macrophages and examined for adipocyte proinflammatory responses and insulin signaling. Compared with their respective levels in controls, WAT iPFK2 amount in HFD-fed mice and WAT SVC PFKFB3 mRNAs in rosiglitazone-treated mice were significantly increased. When the inflammatory responses were analyzed, peritoneal macrophages from PFKFB3-disrputed mice revealed increased proinflammatory activation and decreased anti-inflammatory activation compared with control macrophages. At the whole animal level, hematopoietic cell-specific PFKFB3 disruption enhanced the effects of HFD feeding on promoting WAT inflammation, impairing WAT insulin signaling, and increasing systemic insulin resistance. In vitro, adipocytes co-cultured with PFKFB3-disrupted macrophages revealed increased proinflammatory responses and decreased insulin signaling compared with adipocytes co-cultured with control macrophages. These results suggest that PFKFB3 disruption in hematopoietic cells only exacerbates HFD-induced WAT inflammation and systemic insulin resistance.
Collapse
Affiliation(s)
- Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Guo
- Department of Nutrition, Texas A&M University, College Station, TX, USA
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Shandong, Jinan, China
| | - Hang Xu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Boxiong Jiang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Yina Wang
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiongli Yin
- Department of VIP Medical Service Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tianhao Zhou
- Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - James J Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Shannon Glaser
- Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Fanyin Meng
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Heather Francis
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
16
|
Ghafouri-Fard S, Taheri M. The expression profile and role of non-coding RNAs in obesity. Eur J Pharmacol 2020; 892:173809. [PMID: 33345852 DOI: 10.1016/j.ejphar.2020.173809] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Latest years have experienced a dramatic upsurge in the knowledge about the function of non-coding transcripts in the determination of diverse human phenotypes including obesity. Several miRNAs and lncRNAs participate in the regulation of metabolic pathways leading to obesity. Several lncRNAs such as Mist, lincIRS2, lncRNA-p5549, H19, GAS5 and SNHG9 have been shown to be down-regulated in adipose tissues or other biological samples in the obese human or animal subjects. On the other hand, Meg3, Plnc1, Blnc1, AC092834.1, TINCR and PVT1 are among up-regulated lncRNAs in the obese subjects. Tens of miRNAs have differential expression between obese and non-obese subjects or between mature adipocytes and pre-adipocytes. Understanding the molecular mechanism of involvement of non-coding RNAs in the pathobiology of obesity would simplify design of therapeutic choices for protecting against obesity and its related comorbidities. We explain the available literature on the function of these transcripts in the pathobiology of obesity.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Guo X, Zhu B, Xu H, Li H, Jiang B, Wang Y, Zheng B, Glaser S, Alpini G, Wu C. Adoptive transfer of Pfkfb3-disrupted hematopoietic cells to wild-type mice exacerbates diet-induced hepatic steatosis and inflammation. LIVER RESEARCH 2020; 4:136-144. [PMID: 34336366 PMCID: PMC8320599 DOI: 10.1016/j.livres.2020.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES Hepatic steatosis and inflammation are key characteristics of non-alcoholic fatty liver disease (NAFLD). However, whether and how hepatic steatosis and liver inflammation are differentially regulated remains to be elucidated. Considering that disruption of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (Pfkfb3/iPfk2) dissociates fat deposition and inflammation, the present study examined a role for Pfkfb3/iPfk2 in hematopoietic cells in regulating hepatic steatosis and inflammation in mice. METHODS Pfkfb3-disrupted (Pfkfb3 +/-) mice and wild-type (WT) littermates were fed a high-fat diet (HFD) and examined for NAFLD phenotype. Also, bone marrow cells isolated from Pfkfb3 +/- mice and WT mice were differentiated into macrophages for analysis of macrophage activation status and for bone marrow transplantation (BMT) to generate chimeric (WT/BMT- Pfkfb3 +/-) mice in which Pfkfb3 was disrupted only in hematopoietic cells and control chimeric (WT/BMT-WT) mice. The latter were also fed an HFD and examined for NAFLD phenotype. In vitro, hepatocytes were co-cultured with bone marrow-derived macrophages and examined for hepatocyte fat deposition and proinflammatory responses. RESULTS After the feeding period, HFD-fed Pfkfb3 +/- mice displayed increased severity of liver inflammation in the absence of hepatic steatosis compared with HFD-fed WT mice. When inflammatory activation was analyzed, Pfkfb3 +/- macrophages revealed increased proinflammatory activation and decreased anti-proinflammatory activation. When NAFLD phenotype was analyzed in the chimeric mice, WT/BMT-Pfkfb3 +/- mice displayed increases in the severity of HFD-induced hepatic steatosis and inflammation compared with WT/BMT-WT mice. At the cellular level, hepatocytes co-cultured with Pfkfb3 +/- macrophages revealed increased fat deposition and proinflammatory responses compared with hepatocytes co-cultured with WT macrophages. CONCLUSIONS Pfkfb3 disruption only in hematopoietic cells exacerbates HFD-induced hepatic steatosis and inflammation whereas the Pfkfb3/iPfk2 in nonhematopoietic cells appeared to be needed for HFD feeding to induce hepatic steatosis. As such, the Pfkfb3/iPfk2 plays a unique role in regulating NAFLD pathophysiology.
Collapse
Affiliation(s)
- Xin Guo
- Department of Nutrition, Texas A&M University, College Station, TX, USA,Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bilian Zhu
- Department of Nutrition, Texas A&M University, College Station, TX, USA,Department of VIP Medical Service Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hang Xu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Honggui Li
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Boxiong Jiang
- Department of VIP Medical Service Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yina Wang
- Department of VIP Medical Service Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Benrong Zheng
- Department of VIP Medical Service Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shannon Glaser
- Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN, USA,Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, USA,Corresponding author. Department of Nutrition, Texas A&M University, College Station, TX, USA. (C. Wu)
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Diabetes is a spectrum of clinical manifestations, including latent autoimmune diabetes in adults (LADA). However, it has been questioned whether LADA exists or simply is a group of misclassified type 1 diabetes (T1D) and type 2 diabetes (T2D) patients. This review will provide an updated overview of the genetics of LADA, highlight what genetics tell us about LADA as a diabetes subtype, and point to future directions in the study of LADA. RECENT FINDINGS Recent studies have verified the genetic overlap between LADA and both T1D and T2D and have contributed identification of a novel LADA-specific locus, namely, PFKFB3, and subtype-specific signatures in the HLA region. Genetic risk scores comprising T1D-risk variants have been shown to be a promising tool for discriminating diabetes subtypes and identifying patients rapidly progressing to insulin dependence. Genetic data support the existence of LADA, but further studies are needed to fully determine the place of LADA in the diabetes spectrum.
Collapse
Affiliation(s)
- Mette K Andersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
19
|
A Comprehensive Genome-Wide and Phenome-Wide Examination of BMI and Obesity in a Northern Nevadan Cohort. G3-GENES GENOMES GENETICS 2020; 10:645-664. [PMID: 31888951 PMCID: PMC7003082 DOI: 10.1534/g3.119.400910] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The aggregation of Electronic Health Records (EHR) and personalized genetics leads to powerful discoveries relevant to population health. Here we perform genome-wide association studies (GWAS) and accompanying phenome-wide association studies (PheWAS) to validate phenotype-genotype associations of BMI, and to a greater extent, severe Class 2 obesity, using comprehensive diagnostic and clinical data from the EHR database of our cohort. Three GWASs of 500,000 variants on the Illumina platform of 6,645 Healthy Nevada participants identified several published and novel variants that affect BMI and obesity. Each GWAS was followed with two independent PheWASs to examine associations between extensive phenotypes (incidence of diagnoses, condition, or disease), significant SNPs, BMI, and incidence of extreme obesity. The first GWAS examines associations with BMI in a cohort with no type 2 diabetics, focusing exclusively on BMI. The second GWAS examines associations with BMI in a cohort that includes type 2 diabetics. In the second GWAS, type 2 diabetes is a comorbidity, and thus becomes a covariate in the statistical model. The intersection of significant variants of these two studies is surprising. The third GWAS is a case vs. control study, with cases defined as extremely obese (Class 2 or 3 obesity), and controls defined as participants with BMI between 18.5 and 25. This last GWAS identifies strong associations with extreme obesity, including established variants in the FTO and NEGR1 genes, as well as loci not yet linked to obesity. The PheWASs validate published associations between BMI and extreme obesity and incidence of specific diagnoses and conditions, yet also highlight novel links. This study emphasizes the importance of our extensive longitudinal EHR database to validate known associations and identify putative novel links with BMI and obesity.
Collapse
|
20
|
Hu W, Ding Y, Wang S, Xu L, Yu H. The Construction and Analysis of the Aberrant lncRNA-miRNA-mRNA Network in Adipose Tissue from Type 2 Diabetes Individuals with Obesity. J Diabetes Res 2020; 2020:3980742. [PMID: 32337289 PMCID: PMC7168724 DOI: 10.1155/2020/3980742] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/12/2020] [Accepted: 03/12/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The prevalence of obesity and type 2 diabetes mellitus (T2DM) has become the most serious global public health issue. In recent years, there has been increasing attention to the role of long noncoding RNAs (lncRNAs) in the occurrence and development of obesity and T2DM. The aim of this work was to find new lncRNAs as potential predictive biomarkers or therapeutic targets for obesity and T2DM. METHODS In this study, we identified significant differentially expressed mRNAs (DEmRNAs) and differentially expressed lncRNAs (DElncRNAs) between adipose tissue of individuals with obesity and T2DM and normal adipose tissue (absolute log2FC ≥ 1 and FDR < 0.05). Then, the lncRNA-miRNA interactions predicted by miRcode were further screened with a threshold of MIC > 0.2. Simultaneously, the mRNA-miRNA interactions were explored by miRWalk 2.0. Finally, a ceRNA network consisting of lncRNAs, miRNAs, and mRNAs was established by integrating lncRNA-miRNA interactions and mRNA-miRNA interactions. RESULTS Upon comparing adipose tissue from individuals with obesity and T2DM and normal adipose tissues, 364 significant DEmRNAs, including 140 upregulated and 224 downregulated mRNAs, were identified in GSE104674; in addition, 231 significant DEmRNAs, including 146 upregulated and 85 downregulated mRNAs, were identified in GSE133099. GO and KEGG analyses have shown that downregulated DEmRNAs in GSE104674 and GSE133099 were associated with obesity- and T2DM-related biological pathways, such as lipid metabolism, AMPK signaling, and insulin resistance. Furthermore, 28 significant DElncRNAs, including 14 upregulated and 14 downregulated lncRNAs, were found. Based on the predicted lncRNA-miRNA and mRNA-miRNA relationships, we constructed a competitive endogenous RNA (ceRNA) network, including five lncRNAs, ten miRNAs, and 15 mRNAs. KEGG-GSEA analysis revealed that four lncRNAs (FLG-AS1, SNAI3-AS1, AC008147.0, and LINC02015) in the ceRNA network were related to the biological pathways of metabolic diseases. CONCLUSIONS Through ceRNA network analysis, our study identified four new lncRNAs that may be used as potential biomarkers and therapeutic targets of obesity and T2DM, thus laying a foundation for future clinical studies.
Collapse
Affiliation(s)
- Wei Hu
- Department of Epidemiology and Medical Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yuanlin Ding
- Department of Epidemiology and Medical Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, China
| | - Shu Wang
- Department of Epidemiology and Medical Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, China
| | - Lin Xu
- Department of Epidemiology and Medical Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, China
| | - Haibing Yu
- Department of Epidemiology and Medical Statistics, School of Public Health, Guangdong Medical University, Dongguan, Guangdong, China
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
21
|
Zhou J, Li H, Cai Y, Ma L, Matthews D, Lu B, Zhu B, Chen Y, Qian X, Xiao X, Li Q, Guo S, Huo Y, Zhao L, Tian Y, Li Q, Wu C. Mice lacking adenosine 2A receptor reveal increased severity of MCD-induced NASH. J Endocrinol 2019; 243:JOE-19-0198.R1. [PMID: 31505462 PMCID: PMC7050433 DOI: 10.1530/joe-19-0198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022]
Abstract
Adenosine 2A receptor (A2AR) exerts a protective role in obesity-related non-alcoholic fatty liver disease. Here, we examined whether A2AR protects against non-alcoholic steatohepatitis (NASH). In C57BL/6J mice, feeding a methionine- and choline-deficient diet (MCD) resulted in significant weight loss, overt hepatic steatosis, and massive aggregation of macrophages in the liver compared with mice fed a chow diet. MCD feeding also significantly increased the numbers of A2AR-positive macrophages/Kupffer cells in liver sections although decreasing A2AR amount in liver lysates compared with chow diet feeding. Next, MCD-induced NASH phenotype was examined in A2AR-disrupted mice and control mice. Upon MCD feeding, A2AR-disruptd mice and control mice displayed comparable decreases in body weight and fat mass. However, MCD-fed A2AR-disrupted mice revealed greater liver weight and increased severity of hepatic steatosis compared with MCD-fed control mice. Moreover, A2AR-disupted mice displayed increased severity of MCD-induced liver inflammation, indicated by massive aggregation of macrophages and increased phosphorylation states of Jun-N terminal kinase (JNK) p46 and nuclear factor kappa B (NFκB) p65 and mRNA levels of tumor necrosis factor alpha, interleukin-1 beta, and interleukin-6. In vitro, incubation with MCD-mimicking media increased lipopolysaccharide (LPS)-induced phosphorylation states of JNK p46 and/or NFκB p65 and cytokine mRNAs in control macrophages and RAW264.7 cells, but not primary hepatocytes. Additionally, MCD-mimicking media significantly increased lipopolysaccharide-induced phosphorylation states of p38 and NFκB p65 in A2AR-deficient macrophages, but insignificantly decreased lipopolysaccharide-induced phosphorylation states of JNK p46 and NFκB p65 in A2AR-deficient hepatocytes. Collectively, these results suggest that A2AR disruption exacerbates MCD-induced NASH, which is attributable to, in large part, increased inflammatory responses in macrophages.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Yuli Cai
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
- Department of Endocrinology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, China
| | - Linqiang Ma
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Destiny Matthews
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Bangchao Lu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
- Department of Geriatrics, the Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangshu 211166, USA
| | - Bilian Zhu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
- Department of Endocrinology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Yanming Chen
- Department of Endocrinology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Xiaoxian Qian
- Department of Cardiology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Xiaoqiu Xiao
- Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qifu Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shaodong Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Liang Zhao
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
22
|
RNA-Sequencing Analysis of Paternal Low-Protein Diet-Induced Gene Expression Change in Mouse Offspring Adipocytes. G3-GENES GENOMES GENETICS 2019; 9:2161-2170. [PMID: 31289120 PMCID: PMC6643888 DOI: 10.1534/g3.119.400181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increasing evidence indicates that parental diet affects the metabolism and health of offspring. It is reported that paternal low-protein diet (pLPD) induces glucose intolerance and the expression of genes involved in cholesterol biosynthesis in mouse offspring liver. The aim of the present study was to determine the effect of a pLPD on gene expression in offspring white adipose tissue (WAT), another important tissue for the regulation of metabolism. RNA-seq analysis indicated that pLPD up- and down-regulated 54 and 274 genes, respectively, in offspring WAT. The mRNA expression of many genes involved in lipogenesis was down-regulated by pLPD feeding, which may contribute to metabolic disorder. The expression of carbohydrate response element-binding protein β (ChREBP-β), an important lipogenic transcription factor, was also significantly lower in the WAT of pLPD offspring, which may have mediated the down-regulation of the lipogenic genes. By contrast, the LPD did not affect the expression of lipogenic genes in the WAT of the male progenitor, but increased the expression of lipid oxidation genes, suggesting that a LPD may reduce lipogenesis using different mechanisms in parents and offspring. These findings add to our understanding of how paternal diet can regulate metabolism in their offspring.
Collapse
|
23
|
Min SY, Learnard H, Kant S, Gealikman O, Rojas-Rodriguez R, DeSouza T, Desai A, Keaney JF, Corvera S, Craige SM. Exercise Rescues Gene Pathways Involved in Vascular Expansion and Promotes Functional Angiogenesis in Subcutaneous White Adipose Tissue. Int J Mol Sci 2019; 20:ijms20082046. [PMID: 31027261 PMCID: PMC6515240 DOI: 10.3390/ijms20082046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/05/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023] Open
Abstract
Exercise mitigates chronic diseases such as diabetes, cardiovascular diseases, and obesity; however, the molecular mechanisms governing protection from these diseases are not completely understood. Here we demonstrate that exercise rescues metabolically compromised high fat diet (HFD) fed mice, and reprograms subcutaneous white adipose tissue (scWAT). Using transcriptomic profiling, scWAT was analyzed for HFD gene expression changes that were rescued by exercise. Gene networks involved in vascularization were identified as prominent targets of exercise, which led us to investigate the vasculature architecture and endothelial phenotype. Vascular density in scWAT was found to be compromised in HFD, and exercise rescued this defect. Similarly, angiogenic capacity as measured by ex vivo capillary sprouting was significantly promoted with exercise. Together, these data demonstrate that exercise enhances scWAT vascularization and functional capacity for angiogenesis, and can prevent the detrimental effects of HFD. The improvement in these indices correlates with improvement of whole-body metabolism, suggesting that scWAT vascularization may be a potential therapeutic target for metabolic disease.
Collapse
Affiliation(s)
- So Yun Min
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Heather Learnard
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Olga Gealikman
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Raziel Rojas-Rodriguez
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Anand Desai
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - John F Keaney
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Siobhan M Craige
- Human Nutrition, Food, and Exercise Department, Virginia Tech, Blacksburg, VA 24060, USA.
| |
Collapse
|
24
|
Mahmassani ZS, Reidy PT, McKenzie AI, Stubben C, Howard MT, Drummond MJ. Disuse-induced insulin resistance susceptibility coincides with a dysregulated skeletal muscle metabolic transcriptome. J Appl Physiol (1985) 2019; 126:1419-1429. [PMID: 30763167 DOI: 10.1152/japplphysiol.01093.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Short-term muscle disuse is characterized by skeletal muscle insulin resistance, although this response is divergent across subjects. The mechanisms regulating inactivity-induced insulin resistance between populations that are more or less susceptible to disuse-induced insulin resistance are not known. RNA sequencing was conducted on vastus lateralis muscle biopsies from subjects before and after bed rest (n = 26) to describe the transcriptome of inactivity-induced insulin resistance. Subjects were separated into Low (n = 14) or High (n = 12) Susceptibility Groups based on the magnitude of change in insulin sensitivity after 5 days of bed rest. Both groups became insulin-resistant after bed rest, and there were no differences between groups in nonmetabolic characteristics (body mass, body mass index, fat mass, and lean mass). The High Susceptibility Group had more genes altered >1.5-fold (426 high versus 391 low) and more than twofold (73 high versus 55 low). Twenty-four genes were altered more than twofold in the High Susceptibility Group that did not change in the Low Susceptibility Group. 95 gene changes correlated with the changes in insulin sensitivity; 6 of these genes changed more than twofold in the High Susceptibility Group. Participants in the High Susceptibility Group were uniquely characterized with muscle gene responses described by a decrease in pathways responsible for lipid uptake and oxidation, decreased capacity for triglyceride export (APOB), increased lipogenesis (i.e., PFKFB3, FASN), and increased amino acid export (SLC43A1). These transcriptomic data provide a comprehensive examination of pathways and genes that may be useful biomarkers, or novel targets to offset muscle disuse-induced insulin resistance. NEW & NOTEWORTHY Short-term muscle disuse results in skeletal muscle insulin resistance through mechanisms that are not fully understood. Following a 5-day bed rest intervention, subjects were divided into High and Low Susceptibility Groups to inactivity-induced insulin resistance. This was followed by a genome-wide transcriptional analysis on muscle biopsy samples to gain insight on divergent insulin sensitivity responses. Our primary finding was that the skeletal muscle of subjects who experienced the most inactivity-induced insulin resistance (high susceptibility) was characterized by a decreased preference for lipid oxidation, increased lipogenesis, and increased amino acid export.
Collapse
Affiliation(s)
- Ziad S Mahmassani
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| | - Paul T Reidy
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| | - Alec I McKenzie
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| | - Chris Stubben
- Bioinformatics Shared Resource at the Huntsman Cancer Institute , Salt Lake City, Utah
| | - Michael T Howard
- Department of Genetics, University of Utah , Salt Lake City, Utah
| | - Micah J Drummond
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| |
Collapse
|
25
|
Luo X, Li H, Ma L, Zhou J, Guo X, Woo SL, Pei Y, Knight LR, Deveau M, Chen Y, Qian X, Xiao X, Li Q, Chen X, Huo Y, McDaniel K, Francis H, Glaser S, Meng F, Alpini G, Wu C. Expression of STING Is Increased in Liver Tissues From Patients With NAFLD and Promotes Macrophage-Mediated Hepatic Inflammation and Fibrosis in Mice. Gastroenterology 2018; 155:1971-1984.e4. [PMID: 30213555 PMCID: PMC6279491 DOI: 10.1053/j.gastro.2018.09.010] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/17/2018] [Accepted: 09/04/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Transmembrane protein 173 (TMEM173 or STING) signaling by macrophage activates the type I interferon-mediated innate immune response. The innate immune response contributes to hepatic steatosis and non-alcoholic fatty liver disease (NAFLD). We investigated whether STING regulates diet-induced in hepatic steatosis, inflammation, and liver fibrosis in mice. METHODS Mice with disruption of Tmem173 (STINGgt) on a C57BL/6J background, mice without disruption of this gene (controls), and mice with disruption of Tmem173 only in myeloid cells were fed a standard chow diet, a high-fat diet (HFD; 60% fat calories), or a methionine- and choline-deficient diet (MCD). Liver tissues were collected and analyzed by histology and immunohistochemistry. Bone marrow cells were isolated from mice, differentiated into macrophages, and incubated with 5,6-dimethylxanthenone-4-acetic acid (DMXAA; an activator of STING) or cyclic guanosine monophosphate-adenosine monophosphate (cGAMP). Macrophages or their media were applied to mouse hepatocytes or human hepatic stellate cells (LX2) cells, which were analyzed for cytokine expression, protein phosphorylation, and fat deposition (by oil red O staining after incubation with palmitate). We obtained liver tissues from patients with and without NAFLD and analyzed these by immunohistochemistry. RESULTS Non-parenchymal cells of liver tissues from patients with NAFLD had higher levels of STING than cells of liver tissues from patients without NAFLD. STINGgt mice and mice with disruption only in myeloid cells developed less severe hepatic steatosis, inflammation, and/or fibrosis after the HFD or MCD than control mice. Levels of phosphorylated c-Jun N-terminal kinase and p65 and mRNAs encoding tumor necrosis factor and interleukins 1B and 6 (markers of inflammation) were significantly lower in liver tissues from STINGgt mice vs control mice after the HFD or MCD. Transplantation of bone marrow cells from control mice to STINGgt mice restored the severity of steatosis and inflammation after the HFD. Macrophages from control, but not STINGgt, mice increased markers of inflammation in response to lipopolysaccharide and cGAMP. Hepatocytes and stellate cells cocultured with STINGgt macrophages in the presence of DMXAA or incubated with the medium collected from these macrophages had decreased fat deposition and markers of inflammation compared with hepatocytes or stellate cells incubated with control macrophages. CONCLUSIONS Levels of STING were increased in liver tissues from patients with NAFLD and mice with HFD-induced steatosis. In mice, loss of STING from macrophages decreased the severity of liver fibrosis and the inflammatory response. STING might be a therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Xianjun Luo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Linqiang Ma
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,,Department of Endocrinology, Texas A&M University, College Station, TX 77843, USA,Department of the Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jing Zhou
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Xin Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Shih-Lung Woo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Linda R. Knight
- Department of Radiation Oncology, Veterinary Medical Teaching Hospital, Texas A&M University, College Station, TX 77843, USA
| | - Michael Deveau
- Department of Radiation Oncology, Veterinary Medical Teaching Hospital, Texas A&M University, College Station, TX 77843, USA
| | - Yanming Chen
- Department of Endocrinology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Xiaoxian Qian
- Department of Cardiology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, Texas A&M University, College Station, TX 77843, USA
| | - Qifu Li
- Department of the Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiangbai Chen
- Department of Pathology, Baylor Scott & White Health, College Station, TX 77845; USA
| | - Yuqing Huo
- Department of Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Kelly McDaniel
- Department of Research, Central Texas Veterans Health Care System,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504
| | - Heather Francis
- Department of Research, Central Texas Veterans Health Care System,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504
| | - Shannon Glaser
- Department of Research, Central Texas Veterans Health Care System,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504
| | - Fanyin Meng
- Department of Research, Central Texas Veterans Health Care System
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, Texas; Department of Medical Physiology, Texas A&M University College of Medicine, Temple, Texas.
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas.
| |
Collapse
|
26
|
Pei Y, Li H, Cai Y, Zhou J, Luo X, Ma L, McDaniel K, Zeng T, Chen Y, Qian X, Huo Y, Glaser S, Meng F, Alpini G, Chen L, Wu C. Regulation of adipose tissue inflammation by adenosine 2A receptor in obese mice. J Endocrinol 2018; 239:365-376. [PMID: 30400017 PMCID: PMC6226050 DOI: 10.1530/joe-18-0169] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 09/17/2018] [Indexed: 12/25/2022]
Abstract
Adenosine 2A receptor (A2AR) exerts anti-inflammatory effects. However, the role of A2AR in obesity-associated adipose tissue inflammation remains to be elucidated. The present study examined the expression of A2AR in adipose tissue of mice with diet-induced obesity and determined the effect of A2AR disruption on the status of obesity-associated adipose tissue inflammation. WT C57BL/6J mice and A2AR-disrupted mice were fed a high-fat diet (HFD) for 12 weeks to induce obesity and adipose tissue inflammation. In vitro, bone marrow-derived macrophages from A2AR-disrupted mice and WT control mice were treated with palmitate and examined for macrophage proinflammatory activation. Compared with that of low-fat diet (LFD)-fed WT mice, A2AR expression in adipose tissue of HFD-fed WT mice was increased significantly and was present predominantly in adipose tissue macrophages. The increase in adipose tissue A2AR expression in HFD-fed mice was accompanied with increased phosphorylation states of c-Jun N-terminal kinase 1 p46 and nuclear factor kappa B p65 and mRNA levels of interleukin (Il)-1beta, Il6 and tumor necrosis factor alpha. In A2AR-disrupted mice, HFD feeding induced significant increases in adipose tissue inflammation, indicated by enhanced proinflammatory signaling and increased proinflammatory cytokine expression, and adipose tissue insulin resistance, indicated by a decrease in insulin-stimulated Akt phosphorylation relative to those in WT mice. Lastly, A2AR disruption enhanced palmitate-induced macrophage proinflammatory activation. Taken together, these results suggest that A2AR plays a protective role in obesity-associated adipose tissue inflammation, which is attributable to, in large part, A2AR suppression of macrophage proinflammatory activation.
Collapse
Affiliation(s)
- Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| | - Yuli Cai
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Zhou
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| | - Xianjun Luo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| | - Linqiang Ma
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| | - Kelly McDaniel
- Research, Central Texas Veterans Health Care System, Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Department of Medical Physiology, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanming Chen
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoxian Qian
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Department of Medical Physiology, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Department of Medical Physiology, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Department of Medical Physiology, Texas A&M University College of Medicine, Temple, Texas, USA
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
27
|
Cousminer DL, Ahlqvist E, Mishra R, Andersen MK, Chesi A, Hawa MI, Davis A, Hodge KM, Bradfield JP, Zhou K, Guy VC, Åkerlund M, Wod M, Fritsche LG, Vestergaard H, Snyder J, Højlund K, Linneberg A, Käräjämäki A, Brandslund I, Kim CE, Witte D, Sørgjerd EP, Brillon DJ, Pedersen O, Beck-Nielsen H, Grarup N, Pratley RE, Rickels MR, Vella A, Ovalle F, Melander O, Harris RI, Varvel S, Grill VE, Hakonarson H, Froguel P, Lonsdale JT, Mauricio D, Schloot NC, Khunti K, Greenbaum CJ, Åsvold BO, Yderstræde KB, Pearson ER, Schwartz S, Voight BF, Hansen T, Tuomi T, Boehm BO, Groop L, Leslie RD, Grant SF. First Genome-Wide Association Study of Latent Autoimmune Diabetes in Adults Reveals Novel Insights Linking Immune and Metabolic Diabetes. Diabetes Care 2018; 41:2396-2403. [PMID: 30254083 PMCID: PMC6196829 DOI: 10.2337/dc18-1032] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/26/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Latent autoimmune diabetes in adults (LADA) shares clinical features with both type 1 and type 2 diabetes; however, there is ongoing debate regarding the precise definition of LADA. Understanding its genetic basis is one potential strategy to gain insight into appropriate classification of this diabetes subtype. RESEARCH DESIGN AND METHODS We performed the first genome-wide association study of LADA in case subjects of European ancestry versus population control subjects (n = 2,634 vs. 5,947) and compared against both case subjects with type 1 diabetes (n = 2,454 vs. 968) and type 2 diabetes (n = 2,779 vs. 10,396). RESULTS The leading genetic signals were principally shared with type 1 diabetes, although we observed positive genetic correlations genome-wide with both type 1 and type 2 diabetes. Additionally, we observed a novel independent signal at the known type 1 diabetes locus harboring PFKFB3, encoding a regulator of glycolysis and insulin signaling in type 2 diabetes and inflammation and autophagy in autoimmune disease, as well as an attenuation of key type 1-associated HLA haplotype frequencies in LADA, suggesting that these are factors that distinguish childhood-onset type 1 diabetes from adult autoimmune diabetes. CONCLUSIONS Our results support the need for further investigations of the genetic factors that distinguish forms of autoimmune diabetes as well as more precise classification strategies.
Collapse
Affiliation(s)
- Diana L. Cousminer
- Corresponding authors: Diana L. Cousminer, , and R. David Leslie, , and Struan F.A. Grant,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - R. David Leslie
- Corresponding authors: Diana L. Cousminer, , and R. David Leslie, , and Struan F.A. Grant,
| | - Struan F.A. Grant
- Corresponding authors: Diana L. Cousminer, , and R. David Leslie, , and Struan F.A. Grant,
| |
Collapse
|
28
|
Bartrons R, Rodríguez-García A, Simon-Molas H, Castaño E, Manzano A, Navarro-Sabaté À. The potential utility of PFKFB3 as a therapeutic target. Expert Opin Ther Targets 2018; 22:659-674. [PMID: 29985086 DOI: 10.1080/14728222.2018.1498082] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
INTRODUCTION It has been known for over half a century that tumors exhibit an increased demand for nutrients to fuel their rapid proliferation. Interest in targeting cancer metabolism to treat the disease has been renewed in recent years with the discovery that many cancer-related pathways have a profound effect on metabolism. Considering the recent increase in our understanding of cancer metabolism and the enzymes and pathways involved, the question arises as to whether metabolism is cancer's Achilles heel. Areas covered: This review summarizes the role of 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) in glycolysis, cell proliferation, and tumor growth, discussing PFKFB3 gene and isoenzyme regulation and the changes that occur in cancer and inflammatory diseases. Pharmacological options currently available for selective PFKFB3 inhibition are also reviewed. Expert opinion: PFKFB3 plays an important role in sustaining the development and progression of cancer and might represent an attractive target for therapeutic strategies. Nevertheless, clinical trials are needed to follow up on the promising results from preclinical studies with PFKFB3 inhibitors. Combination therapies with PFKFB3 inhibitors, chemotherapeutic drugs, or radiotherapy might improve the efficacy of cancer treatments targeting PFKFB3.
Collapse
Affiliation(s)
- Ramon Bartrons
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Ana Rodríguez-García
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Helga Simon-Molas
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Esther Castaño
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Anna Manzano
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| | - Àurea Navarro-Sabaté
- a Unitat de Bioquímica, Departament de Ciències Fisiològiques , Universitat de Barcelona, IDIBELL , Catalunya , Spain
| |
Collapse
|
29
|
Minchenko OH, Bashta YM, Minchenko DO, Ratushna OO. Glucose tolerance in obese men is associated with dysregulation of some angiogenesis-related gene expressions in subcutaneous adipose tissue. ACTA ACUST UNITED AC 2018. [PMID: 29537219 DOI: 10.15407/fz62.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Obesity and its metabolic complications are one of the most profound public health problems and result from interactions between genes and environmental. The development of obesity is tightly connected with dysregulation of intrinsic gene expression mechanisms controlling majority of metabolic processes, which are essential for regulation many physiological functions, including insulin sensitivity, cellular proliferation and angiogenesis. Our objective was to evaluate if expression of angiogenesis related genes VEGF-A, CYR61, PDGFC, FGF1, FGF2, FGFR2, FGFRL1, E2F8, BAI2, HIF1A, and EPAS1 at mRNA level in adipose tissue could participate in the development of obesity and metabolic complications. We have shown that expression level of VEGF-A, PDGFC, FGF2, and FGFRL1 genes is decreased in adipose tissue of obese men with normal glucose tolerance (NGT) versus a group of control subjects. At the same time, in this group of obese individuals a significant up-regulation of CYR61, FGF1, FGFR2, E2F8, BAI2, and HIF1A gene expressions was observed. Impaired glucose tolerance (IGT) in obese patients associates with down-regulation of CYR61 and FGFR2 mRNA and up-regulations of E2F8, FGF1, FGF2, VEGF-A and its splice variant 189 mRNA expressions in adipose tissue versus obese (NGT) individuals. Thus, our data demonstrate that the expression of almost all studied genes is affected in subcutaneous adipose tissue of obese individuals with NGT and that glucose intolerance is associated with gene-specific changes in the expression of E2F8, FGF1, FGF2, VEGF-A, CYR61 and FGFR2 mRNAs. The data presented here provides evidence that VEGF-A, CYR61, PDGFC, FGF1, FGF2, FGFR2, FGFRL1, E2F8, BAI2, and HIF1A genes are possibly involved in the development of obesity and its complications.
Collapse
MESH Headings
- Adult
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Case-Control Studies
- Cysteine-Rich Protein 61/genetics
- Cysteine-Rich Protein 61/metabolism
- Fibroblast Growth Factor 1/genetics
- Fibroblast Growth Factor 1/metabolism
- Fibroblast Growth Factor 2/genetics
- Fibroblast Growth Factor 2/metabolism
- Gene Expression Regulation
- Glucose/metabolism
- Glucose Intolerance/genetics
- Glucose Intolerance/metabolism
- Glucose Intolerance/pathology
- Glucose Tolerance Test
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Insulin/metabolism
- Insulin Resistance
- Lymphokines/genetics
- Lymphokines/metabolism
- Male
- Middle Aged
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Obesity/genetics
- Obesity/metabolism
- Obesity/pathology
- Platelet-Derived Growth Factor/genetics
- Platelet-Derived Growth Factor/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 5/genetics
- Receptor, Fibroblast Growth Factor, Type 5/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Subcutaneous Fat/blood supply
- Subcutaneous Fat/metabolism
- Subcutaneous Fat/pathology
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
|
30
|
Integration of flux measurements to resolve changes in anabolic and catabolic metabolism in cardiac myocytes. Biochem J 2017; 474:2785-2801. [PMID: 28706006 PMCID: PMC5545928 DOI: 10.1042/bcj20170474] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022]
Abstract
Although ancillary pathways of glucose metabolism are critical for synthesizing cellular building blocks and modulating stress responses, how they are regulated remains unclear. In the present study, we used radiometric glycolysis assays, [13C6]-glucose isotope tracing, and extracellular flux analysis to understand how phosphofructokinase (PFK)-mediated changes in glycolysis regulate glucose carbon partitioning into catabolic and anabolic pathways. Expression of kinase-deficient or phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase in rat neonatal cardiomyocytes co-ordinately regulated glycolytic rate and lactate production. Nevertheless, in all groups, >40% of glucose consumed by the cells was unaccounted for via catabolism to pyruvate, which suggests entry of glucose carbons into ancillary pathways branching from metabolites formed in the preparatory phase of glycolysis. Analysis of 13C fractional enrichment patterns suggests that PFK activity regulates glucose carbon incorporation directly into the ribose and the glycerol moieties of purines and phospholipids, respectively. Pyrimidines, UDP-N-acetylhexosamine, and the fatty acyl chains of phosphatidylinositol and triglycerides showed lower 13C incorporation under conditions of high PFK activity; the isotopologue 13C enrichment pattern of each metabolite indicated limitations in mitochondria-engendered aspartate, acetyl CoA and fatty acids. Consistent with this notion, high glycolytic rate diminished mitochondrial activity and the coupling of glycolysis to glucose oxidation. These findings suggest that a major portion of intracellular glucose in cardiac myocytes is apportioned for ancillary biosynthetic reactions and that PFK co-ordinates the activities of the pentose phosphate, hexosamine biosynthetic, and glycerolipid synthesis pathways by directly modulating glycolytic intermediate entry into auxiliary glucose metabolism pathways and by indirectly regulating mitochondrial cataplerosis.
Collapse
|
31
|
Guo X, Shu C, Li H, Pei Y, Woo SL, Zheng J, Liu M, Xu H, Botchlett R, Guo T, Cai Y, Gao X, Zhou J, Chen L, Li Q, Xiao X, Xie L, Zhang KK, Ji JY, Huo Y, Meng F, Alpini G, Li P, Wu C. Cyclic GMP-AMP Ameliorates Diet-induced Metabolic Dysregulation and Regulates Proinflammatory Responses Distinctly from STING Activation. Sci Rep 2017; 7:6355. [PMID: 28743914 PMCID: PMC5526935 DOI: 10.1038/s41598-017-05884-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/26/2017] [Indexed: 01/22/2023] Open
Abstract
Endogenous cyclic GMP-AMP (cGAMP) binds and activates STING to induce type I interferons. However, whether cGAMP plays any roles in regulating metabolic homeostasis remains unknown. Here we show that exogenous cGAMP ameliorates obesity-associated metabolic dysregulation and uniquely alters proinflammatory responses. In obese mice, treatment with cGAMP significantly decreases diet-induced proinflammatory responses in liver and adipose tissues and ameliorates metabolic dysregulation. Strikingly, cGAMP exerts cell-type-specific anti-inflammatory effects on macrophages, hepatocytes, and adipocytes, which is distinct from the effect of STING activation by DMXAA on enhancing proinflammatory responses. While enhancing insulin-stimulated Akt phosphorylation in hepatocytes and adipocytes, cGAMP weakens the effects of glucagon on stimulating hepatocyte gluconeogenic enzyme expression and glucose output and blunts palmitate-induced hepatocyte fat deposition in an Akt-dependent manner. Taken together, these results suggest an essential role for cGAMP in linking innate immunity and metabolic homeostasis, indicating potential applications of cGAMP in treating obesity-associated inflammatory and metabolic diseases.
Collapse
Affiliation(s)
- Xin Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Chang Shu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Shih-Lung Woo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Juan Zheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Mengyang Liu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Hang Xu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Rachel Botchlett
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Ting Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Yuli Cai
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Xinsheng Gao
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, Texas, 77843, USA
| | - Jing Zhou
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Lu Chen
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Qifu Li
- Department of Endocrinology and the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoqiu Xiao
- Department of Endocrinology and the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,The Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Linglin Xie
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Ke K Zhang
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Jun-Yuan Ji
- Department of Endocrinology and the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.,Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Fanyin Meng
- Departments of Medical Physiology and Medicine, Texas A&M University Health Science Center, Temple, TX, 76504, USA
| | - Gianfranco Alpini
- Departments of Medical Physiology and Medicine, Texas A&M University Health Science Center, Temple, TX, 76504, USA
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA.
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
32
|
Qi T, Chen Y, Li H, Pei Y, Woo SL, Guo X, Zhao J, Qian X, Awika J, Huo Y, Wu C. A role for PFKFB3/iPFK2 in metformin suppression of adipocyte inflammatory responses. J Mol Endocrinol 2017; 59:49-59. [PMID: 28559290 PMCID: PMC5512603 DOI: 10.1530/jme-17-0066] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 12/16/2022]
Abstract
Metformin improves obesity-associated metabolic dysregulation, but has controversial effects on adipose tissue inflammation. The objective of the study is to examine the direct effect of metformin on adipocyte inflammatory responses and elucidate the underlying mechanisms. Adipocytes were differentiated from 3T3-L1 cells and treated with metformin at various doses and for different time periods. The treated cells were examined for the proinflammatory responses, as well as the phosphorylation states of AMPK and the expression of PFKFB3/iPFK2. In addition, PFKFB3/iPFK2-knockdown adipocytes were treated with metformin and examined for changes in the proinflammatory responses. The following results were obtained from the study. Treatment of adipocytes with metformin decreased the effects of lipopolysaccharide on inducing the phosphorylation states of JNK p46 and on increasing the mRNA levels of IL-1β and TNFα. In addition, treatment with metformin increased the expression of PFKFB3/iPFK2, but failed to significantly alter the phosphorylation states of AMPK. In PFKFB3/iPFK2-knockdown adipocytes, treatment with metformin did not suppress the proinflammatory responses as did it in control adipocytes. In conclusion, metformin has a direct effect on suppressing adipocyte proinflammatory responses in an AMPK-independent manner. Also, metformin increases adipocyte expression of PFKFB3/iPFK2, which is involved in the anti-inflammatory effect of metformin.
Collapse
Affiliation(s)
- Ting Qi
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Yanming Chen
- Department of Endocrinologythe Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Honggui Li
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Ya Pei
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Shih-Lung Woo
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Xin Guo
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Jiajia Zhao
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Xiaoxian Qian
- Department of Cardiologythe Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Joseph Awika
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| | - Yuqing Huo
- Vascular Biology CenterDepartment of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, USA
- Drug Discovery CenterKey Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Chaodong Wu
- Department of Nutrition and Food ScienceTexas A&M University, College Station, USA
| |
Collapse
|
33
|
Longo R, Ferrari A, Zocchi M, Crestani M. Of mice and humans through the looking glass: "reflections" on epigenetics of lipid metabolism. Mol Aspects Med 2017; 54:16-27. [PMID: 28119071 DOI: 10.1016/j.mam.2017.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/18/2017] [Accepted: 01/21/2017] [Indexed: 10/20/2022]
Abstract
Over the past decade, epigenetics has emerged as a new layer of regulation of gene expression. Several investigations demonstrated that nutrition and lifestyle regulate lipid metabolism by influencing epigenomic remodeling. Studies on animal models highlighted the role of epigenome modifiers in specific metabolic contexts and established clear links between dysregulation of epigenetic mechanisms and metabolic dysfunction. The relevance of findings in animal models has been translated to humans, as epigenome-wide association studies (EWAS) deeply investigated the relationship between lifestyle and epigenetics in human populations. In this review, we will provide an outlook of recent studies addressing the link between epigenetics and lipid metabolism, by comparing results obtained in animal models and in human subjects.
Collapse
Affiliation(s)
- Raffaella Longo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti, 9, 20133 Milano, Italy.
| | - Alessandra Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti, 9, 20133 Milano, Italy.
| | - Monica Zocchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti, 9, 20133 Milano, Italy.
| | - Maurizio Crestani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti, 9, 20133 Milano, Italy.
| |
Collapse
|
34
|
Arner P, Sahlqvist AS, Sinha I, Xu H, Yao X, Waterworth D, Rajpal D, Loomis AK, Freudenberg JM, Johnson T, Thorell A, Näslund E, Ryden M, Dahlman I. The epigenetic signature of systemic insulin resistance in obese women. Diabetologia 2016; 59:2393-2405. [PMID: 27535281 PMCID: PMC5506095 DOI: 10.1007/s00125-016-4074-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Insulin resistance (IR) links obesity to type 2 diabetes. The aim of this study was to explore whether white adipose tissue (WAT) epigenetic dysregulation is associated with systemic IR by genome-wide CG dinucleotide (CpG) methylation and gene expression profiling in WAT from insulin-resistant and insulin-sensitive women. A secondary aim was to determine whether the DNA methylation signature in peripheral blood mononuclear cells (PBMCs) reflects WAT methylation and, if so, can be used as a marker for systemic IR. METHODS From 220 obese women, we selected a total of 80 individuals from either of the extreme ends of the distribution curve of HOMA-IR, an indirect measure of systemic insulin sensitivity. Genome-wide transcriptome and DNA CpG methylation profiling by array was performed on subcutaneous (SAT) and visceral (omental) adipose tissue (VAT). CpG methylation in PBMCs was assayed in the same cohort. RESULTS There were 647 differentially expressed genes (false discovery rate [FDR] 10%) in SAT, all of which displayed directionally consistent associations in VAT. This suggests that IR is associated with dysregulated expression of a common set of genes in SAT and VAT. The average degree of DNA methylation did not differ between the insulin-resistant and insulin-sensitive group in any of the analysed tissues/cells. There were 223 IR-associated genes in SAT containing a total of 336 nominally significant differentially methylated sites (DMS). The 223 IR-associated genes were over-represented in pathways related to integrin cell surface interactions and insulin signalling and included COL5A1, GAB1, IRS2, PFKFB3 and PTPRJ. In VAT there were a total of 51 differentially expressed genes (FDR 10%); 18 IR-associated genes contained a total of 29 DMS. CONCLUSIONS/INTERPRETATION In individuals discordant for insulin sensitivity, the average DNA CpG methylation in SAT and VAT is similar, although specific genes, particularly in SAT, display significantly altered expression and DMS in IR, possibly indicating that epigenetic regulation of these genes influences metabolism.
Collapse
Affiliation(s)
- Peter Arner
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, C2:94, Huddinge, S-141 86, Stockholm, Sweden
| | | | - Indranil Sinha
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Huan Xu
- GlaxoSmithKline R&D, Research Triangle Park, NC, USA
| | - Xiang Yao
- Computational and Systems Biology, Discovery Sciences, Janssen Pharmaceutical, Research & Development, LLC, San Diego, CA, USA
| | | | | | | | | | | | - Anders Thorell
- Department of Surgery, Ersta Hospital, Stockholm, Sweden
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Danderyd, Sweden
| | - Erik Näslund
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Danderyd, Sweden
| | - Mikael Ryden
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, C2:94, Huddinge, S-141 86, Stockholm, Sweden
| | - Ingrid Dahlman
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, C2:94, Huddinge, S-141 86, Stockholm, Sweden.
| |
Collapse
|
35
|
Glucose and Palmitate Differentially Regulate PFKFB3/iPFK2 and Inflammatory Responses in Mouse Intestinal Epithelial Cells. Sci Rep 2016; 6:28963. [PMID: 27387960 PMCID: PMC4937440 DOI: 10.1038/srep28963] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/07/2016] [Indexed: 12/21/2022] Open
Abstract
The gene PFKFB3 encodes for inducible 6-phosphofructo-2-kinase, a glycolysis-regulatory enzyme that protects against diet-induced intestine inflammation. However, it is unclear how nutrient overload regulates PFKFB3 expression and inflammatory responses in intestinal epithelial cells (IECs). In the present study, primary IECs were isolated from small intestine of C57BL/6J mice fed a low-fat diet (LFD) or high-fat diet (HFD) for 12 weeks. Additionally, CMT-93 cells, a cell line for IECs, were cultured in low glucose (LG, 5.5 mmol/L) or high glucose (HG, 27.5 mmol/L) medium and treated with palmitate (50 μmol/L) or bovine serum albumin (BSA) for 24 hr. These cells were analyzed for PFKFB3 and inflammatory markers. Compared with LFD, HFD feeding decreased IEC PFKFB3 expression and increased IEC proinflammatory responses. In CMT-93 cells, HG significantly increased PFKFB3 expression and proinflammatory responses compared with LG. Interestingly, palmitate decreased PFKFB3 expression and increased proinflammatory responses compared with BSA, regardless of glucose concentrations. Furthermore, HG significantly increased PFKFB3 promoter transcription activity compared with LG. Upon PFKFB3 overexpression, proinflammatory responses in CMT-93 cells were decreased. Taken together, these results indicate that in IECs glucose stimulates PFKFB3 expression and palmitate contributes to increased proinflammatory responses. Therefore, PFKFB3 regulates IEC inflammatory status in response to macronutrients.
Collapse
|
36
|
Kim SM, Neuendorff N, Chapkin RS, Earnest DJ. Role of Inflammatory Signaling in the Differential Effects of Saturated and Poly-unsaturated Fatty Acids on Peripheral Circadian Clocks. EBioMedicine 2016; 7:100-11. [PMID: 27322464 PMCID: PMC4913702 DOI: 10.1016/j.ebiom.2016.03.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/23/2016] [Accepted: 03/23/2016] [Indexed: 12/12/2022] Open
Abstract
Inflammatory signaling may play a role in high-fat diet (HFD)-related circadian clock disturbances that contribute to systemic metabolic dysregulation. Therefore, palmitate, the prevalent proinflammatory saturated fatty acid (SFA) in HFD and the anti-inflammatory, poly-unsaturated fatty acid (PUFA), docosahexaenoic acid (DHA), were analyzed for effects on circadian timekeeping and inflammatory responses in peripheral clocks. Prolonged palmitate, but not DHA, exposure increased the period of fibroblast Bmal1-dLuc rhythms. Acute palmitate treatment produced phase shifts of the Bmal1-dLuc rhythm that were larger in amplitude as compared to DHA. These phase-shifting effects were time-dependent and contemporaneous with rhythmic changes in palmitate-induced inflammatory responses. Fibroblast and differentiated adipocyte clocks exhibited cell-specific differences in the time-dependent nature of palmitate-induced shifts and inflammation. DHA and other inhibitors of inflammatory signaling (AICAR, cardamonin) repressed palmitate-induced proinflammatory responses and phase shifts of the fibroblast clock, suggesting that SFA-mediated inflammatory signaling may feed back to modulate circadian timekeeping in peripheral clocks. The saturated fatty acid (SFA) palmitate differentially modulates the circadian timekeeping mechanism in peripheral clocks; Palmitate induces time-dependent phase shifts that coincide with its rhythmic induction of inflammatory signaling; Time-dependent nature of the palmitate-induced phase shifts and inflammatory signaling is cell specific; Inhibitors of inflammatory signaling repress the proinflammatory and phase shifting effects of palmitate; Inflammatory signaling plays a role in the mechanism by which palmitate alters circadian timekeeping in peripheral clocks.
Circadian or 24-hour clocks throughout the body mediate the local temporal coordination of tissue- or cell-specific processes necessary for normal inflammatory responses and metabolic homeostasis. Dysregulation of peripheral clocks and their timekeeping function contribute to obesity-related metabolic disorders (e.g., type 2 diabetes). Our data unveil a novel mechanism by which mutual interactions between peripheral clocks and inflammatory signaling pathways dysregulate circadian timekeeping, and exacerbate proinflammatory responses to saturated fatty acids. These studies will guide the development of chronotherapeutic drug and/or dietary omega-3 fatty acid treatments for managing and preventing metabolic disorders and other inflammation-related pathologies (e.g., cardiovascular disease, stroke, arthritis).
Collapse
Affiliation(s)
- Sam-Moon Kim
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA; Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843-3258, USA
| | - Nichole Neuendorff
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807-3260, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition & Complex Diseases, Center for Translational Environmental Health Research, Texas A&M University, College Station, TX 77843-2253, USA; Department of Nutrition & Food Science, Texas A&M University, College Station, TX 77843-2253, USA; Department of Microbial Pathogenesis and Immunology, Texas A&M University System Health Science Center, College Station, TX 77807-3260, USA.
| | - David J Earnest
- Department of Biology, Texas A&M University, College Station, TX 77843-3258, USA; Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843-3258, USA; Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807-3260, USA.
| |
Collapse
|
37
|
Oestrogen exerts anti-inflammation via p38 MAPK/NF-κB cascade in adipocytes. Obes Res Clin Pract 2016; 10:633-641. [PMID: 27004692 DOI: 10.1016/j.orcp.2016.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 02/24/2016] [Accepted: 02/29/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Oestrogen has anti-inflammatory property in obesity. However, the mechanism is still not defined. OBJECTIVE To investigate the effect of oestrogen on LPS-induced monocyte chemoattractant protein-1 (MCP-1) production in adipocytes. METHODS Lipopolysaccharides (LPS) was used to imitate inflammatory responses and monocyte chemotactic protein-1 (MCP-1) was selected as an inflammatory marker to observe. 17β-Estradiol (E2), SB203580 (SB), pyrrolidine dithiocarbamate (PDTC), pertussis toxin (PTX), wortmannin (WM), p65 siRNA and p38 MAPK siRNA were pre-treated respectively or together in LPS-induced MCP-1. Then p38 MAPK and NF-κB cascade were silenced successively to observe the change of each other. Lastly, oestrogen receptor (ER) α agonist, ERβ agonist and ER antagonist were utilised. RESULTS LPS-induced MCP-1 largely impaired by pre-treatment with E2, SB, PDTC or silencing NF-κB subunit. E2 inhibited LPS-induced MCP-1 in a time- and dose-dependent manner, which was related to the suppression of p65 translocation to nucleus. Furthermore, LPS rapidly activated p38 MAPK, while E2 markedly inhibited this activation. It markedly attenuated LPS-stimulated p65 translocation to nucleus and MCP-1 production by transfecting with p38 MAPK siRNA or using p38 MAPK inhibitor. The oestrogen's inhibitory effect was mimicked by the ERα agonist, but not by the ERβ agonist. The inhibition of E2 on p38 MAPK phosphorylation was prevented by ER antagonist. CONCLUSIONS E2 inhibits LPS-stimulated MCP-1 in adipocytes. This effect is related to the inhibition of p38 MAPK/NF-κB cascade, and ERα appears to be the dominant ER subtype in these events.
Collapse
|
38
|
Berberine Ameliorates Hepatic Steatosis and Suppresses Liver and Adipose Tissue Inflammation in Mice with Diet-induced Obesity. Sci Rep 2016; 6:22612. [PMID: 26936230 PMCID: PMC4776174 DOI: 10.1038/srep22612] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/17/2016] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence demonstrates that berberine (BBR) is beneficial for obesity-associated non-alcoholic fatty liver disease (NAFLD). However, it remains to be elucidated how BBR improves aspects of NAFLD. Here we revealed an AMP-activated protein kinase (AMPK)-independent mechanism for BBR to suppress obesity-associated inflammation and improve hepatic steatosis. In C57BL/6J mice fed a high-fat diet (HFD), treatment with BBR decreased inflammation in both the liver and adipose tissue as indicated by reduction of the phosphorylation state of JNK1 and the mRNA levels of proinflammatory cytokines. BBR treatment also decreased hepatic steatosis, as well as the expression of acetyl-CoA carboxylase and fatty acid synthase. Interestingly, treatment with BBR did not significantly alter the phosphorylation state of AMPK in both the liver and adipose tissue of HFD-fed mice. Consistently, BBR treatment significantly decreased the phosphorylation state of JNK1 in both hepatoma H4IIE cells and mouse primary hepatocytes in both dose-dependent and time-dependent manners, which was independent of AMPK phosphorylation. BBR treatment also caused a decrease in palmitate-induced fat deposition in primary mouse hepatocytes. Taken together, these results suggest that BBR actions on improving aspects of NAFLD are largely attributable to BBR suppression of inflammation, which is independent of AMPK.
Collapse
|
39
|
Mahesan AM, Ogunyemi D, Kim E, Paul ABM, Chen YDI. Insulin Resistance in Pregnancy Is Correlated with Decreased Insulin Receptor Gene Expression in Omental Adipose: Insulin Sensitivity and Adipose Tissue Gene Expression in Normal Pregnancy. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/jdm.2016.61011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Ramírez-Espinosa JJ, González-Dávalos L, Shimada A, Piña E, Varela-Echavarria A, Mora O. Bovine (Bos taurus) Bone Marrow Mesenchymal Cell Differentiation to Adipogenic and Myogenic Lineages. Cells Tissues Organs 2015; 201:51-64. [PMID: 26565958 DOI: 10.1159/000440878] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2015] [Indexed: 11/19/2022] Open
Abstract
PURPOSE We evaluated the effect of peroxisome proliferator-activated receptor (PPAR) agonists on the differentiation and metabolic features of bovine bone marrow-derived mesenchymal cells induced to adipogenic or myogenic lineages. METHODS Cells isolated from 7-day-old calves were cultured in basal medium (BM). For adipogenic differentiation, cells were cultured for one passage in BM and then transferred to a medium supplemented with either rosiglitazone, telmisartan, sirtinol or conjugated c-9, t-11 linoleic acid; for myogenic differentiation, third-passage cells were added with either bezafibrate, telmisartan or sirtinol. The expression of PPARx03B3; (an adipogenic differentiation marker), myosin heavy chain (MyHC; a myogenic differentiation marker) and genes related to energy metabolism were measured by quantitative real-time PCR in a completely randomized design. RESULTS For adipogenic differentiation, 20 µM telmisartan showed the highest PPARx03B3; expression (15.58 ± 0.62-fold, p < 0.0001), and differences in the expression of energy metabolism-related genes were found for hexokinase II, phosphofructokinase, adipose triglyceride lipase, acetyl-CoA carboxylase α(ACACα) and fatty acid synthase (p < 0.001), but not for ACACβ (p = 0.4275). For myogenic differentiation, 200 µM bezafibrate showed the highest MyHC expression (73.98 ± 11.79-fold), and differences in the expression of all energy metabolism-related genes were found (p < 0.05). CONCLUSIONS Adipocyte and myocyte differentiation are enhanced with telmisartan and bezafibrate, respectively, and energy uptake, storage and mobilization are improved with both.
Collapse
Affiliation(s)
- Jesus J Ramírez-Espinosa
- Programa de Posgrado en Ciencias de la Produccix00F3;n y de la Salud Animal, Universidad Nacional Autx00F3;noma de Mx00E9;xico (UNAM), Mexico City, Mexico
| | | | | | | | | | | |
Collapse
|
41
|
Trefely S, Khoo PS, Krycer JR, Chaudhuri R, Fazakerley DJ, Parker BL, Sultani G, Lee J, Stephan JP, Torres E, Jung K, Kuijl C, James DE, Junutula JR, Stöckli J. Kinome Screen Identifies PFKFB3 and Glucose Metabolism as Important Regulators of the Insulin/Insulin-like Growth Factor (IGF)-1 Signaling Pathway. J Biol Chem 2015; 290:25834-46. [PMID: 26342081 DOI: 10.1074/jbc.m115.658815] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Indexed: 01/02/2023] Open
Abstract
The insulin/insulin-like growth factor (IGF)-1 signaling pathway (ISP) plays a fundamental role in long term health in a range of organisms. Protein kinases including Akt and ERK are intimately involved in the ISP. To identify other kinases that may participate in this pathway or intersect with it in a regulatory manner, we performed a whole kinome (779 kinases) siRNA screen for positive or negative regulators of the ISP, using GLUT4 translocation to the cell surface as an output for pathway activity. We identified PFKFB3, a positive regulator of glycolysis that is highly expressed in cancer cells and adipocytes, as a positive ISP regulator. Pharmacological inhibition of PFKFB3 suppressed insulin-stimulated glucose uptake, GLUT4 translocation, and Akt signaling in 3T3-L1 adipocytes. In contrast, overexpression of PFKFB3 in HEK293 cells potentiated insulin-dependent phosphorylation of Akt and Akt substrates. Furthermore, pharmacological modulation of glycolysis in 3T3-L1 adipocytes affected Akt phosphorylation. These data add to an emerging body of evidence that metabolism plays a central role in regulating numerous biological processes including the ISP. Our findings have important implications for diseases such as type 2 diabetes and cancer that are characterized by marked disruption of both metabolism and growth factor signaling.
Collapse
Affiliation(s)
- Sophie Trefely
- From the Garvan Institute of Medical Research, Sydney 2010 NSW, Australia
| | - Poh-Sim Khoo
- From the Garvan Institute of Medical Research, Sydney 2010 NSW, Australia, Genentech Inc., South San Francisco, California 94080
| | - James R Krycer
- the Charles Perkins Centre, School of Molecular Bioscience, University of Sydney, Sydney 2006 NSW, Australia, and
| | - Rima Chaudhuri
- the Charles Perkins Centre, School of Molecular Bioscience, University of Sydney, Sydney 2006 NSW, Australia, and
| | - Daniel J Fazakerley
- the Charles Perkins Centre, School of Molecular Bioscience, University of Sydney, Sydney 2006 NSW, Australia, and
| | - Benjamin L Parker
- the Charles Perkins Centre, School of Molecular Bioscience, University of Sydney, Sydney 2006 NSW, Australia, and
| | - Ghazal Sultani
- From the Garvan Institute of Medical Research, Sydney 2010 NSW, Australia
| | - James Lee
- Genentech Inc., South San Francisco, California 94080
| | | | - Eric Torres
- Genentech Inc., South San Francisco, California 94080
| | - Kenneth Jung
- Genentech Inc., South San Francisco, California 94080
| | | | - David E James
- the Charles Perkins Centre, School of Molecular Bioscience, University of Sydney, Sydney 2006 NSW, Australia, and the Sydney Medical School, University of Sydney, Sydney 2006 NSW, Australia
| | | | - Jacqueline Stöckli
- the Charles Perkins Centre, School of Molecular Bioscience, University of Sydney, Sydney 2006 NSW, Australia, and
| |
Collapse
|
42
|
Hansen JS, Zhao X, Irmler M, Liu X, Hoene M, Scheler M, Li Y, Beckers J, Hrabĕ de Angelis M, Häring HU, Pedersen BK, Lehmann R, Xu G, Plomgaard P, Weigert C. Type 2 diabetes alters metabolic and transcriptional signatures of glucose and amino acid metabolism during exercise and recovery. Diabetologia 2015; 58:1845-54. [PMID: 26067360 DOI: 10.1007/s00125-015-3584-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/13/2015] [Indexed: 12/24/2022]
Abstract
AIMS/HYPOTHESIS The therapeutic benefit of physical activity to prevent and treat type 2 diabetes is commonly accepted. However, the impact of the disease on the acute metabolic response is less clear. To this end, we investigated the effect of type 2 diabetes on exercise-induced plasma metabolite changes and the muscular transcriptional response using a complementary metabolomics/transcriptomics approach. METHODS We analysed 139 plasma metabolites and hormones at nine time points, and whole genome expression in skeletal muscle at three time points, during a 60 min bicycle ergometer exercise and a 180 min recovery phase in type 2 diabetic patients and healthy controls matched for age, percentage body fat and maximal oxygen consumption (VO2). RESULTS Pathway analysis of differentially regulated genes upon exercise revealed upregulation of regulators of GLUT4 (SLC2A4RG, FLOT1, EXOC7, RAB13, RABGAP1 and CBLB), glycolysis (HK2, PFKFB1, PFKFB3, PFKM, FBP2 and LDHA) and insulin signal mediators in diabetic participants compared with controls. Notably, diabetic participants had normalised rates of lactate and insulin levels, and of glucose appearance and disappearance, after exercise. They also showed an exercise-induced compensatory regulation of genes involved in biosynthesis and metabolism of amino acids (PSPH, GATM, NOS1 and GLDC), which responded to differences in the amino acid profile (consistently lower plasma levels of glycine, cysteine and arginine). Markers of fat oxidation (acylcarnitines) and lipolysis (glycerol) did not indicate impaired metabolic flexibility during exercise in diabetic participants. CONCLUSIONS/INTERPRETATION Type 2 diabetic individuals showed specific exercise-regulated gene expression. These data provide novel insight into potential mechanisms to ameliorate the disturbed glucose and amino acid metabolism associated with type 2 diabetes.
Collapse
Affiliation(s)
- Jakob S Hansen
- Centre of Inflammation and Metabolism, Centre for Physical Activity Research, Department of Infectious Diseases, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zheng J, Woo SL, Hu X, Botchlett R, Chen L, Huo Y, Wu C. Metformin and metabolic diseases: a focus on hepatic aspects. Front Med 2015; 9:173-86. [PMID: 25676019 PMCID: PMC4567274 DOI: 10.1007/s11684-015-0384-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/24/2014] [Indexed: 12/25/2022]
Abstract
Metformin has been widely used as a first-line anti-diabetic medicine for the treatment of type 2 diabetes (T2D). As a drug that primarily targets the liver, metformin suppresses hepatic glucose production (HGP), serving as the main mechanism by which metformin improves hyperglycemia of T2D. Biochemically, metformin suppresses gluconeogenesis and stimulates glycolysis. Metformin also inhibits glycogenolysis, which is a pathway that critically contributes to elevated HGP. While generating beneficial effects on hyperglycemia, metformin also improves insulin resistance and corrects dyslipidemia in patients with T2D. These beneficial effects of metformin implicate a role for metformin in managing non-alcoholic fatty liver disease. As supported by the results from both human and animal studies, metformin improves hepatic steatosis and suppresses liver inflammation. Mechanistically, the beneficial effects of metformin on hepatic aspects are mediated through both adenosine monophosphate-activated protein kinase (AMPK)-dependent and AMPK-independent pathways. In addition, metformin is generally safe and may also benefit patients with other chronic liver diseases.
Collapse
Affiliation(s)
- Juan Zheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shih-Lung Woo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Xiang Hu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rachel Botchlett
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuqing Huo
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
44
|
Affiliation(s)
- Xinghui Sun
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mark W Feinberg
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
45
|
Xu H, Li H, Woo SL, Kim SM, Shende VR, Neuendorff N, Guo X, Guo T, Qi T, Pei Y, Zhao Y, Hu X, Zhao J, Chen L, Chen L, Ji JY, Alaniz RC, Earnest DJ, Wu C. Myeloid cell-specific disruption of Period1 and Period2 exacerbates diet-induced inflammation and insulin resistance. J Biol Chem 2014; 289:16374-88. [PMID: 24770415 DOI: 10.1074/jbc.m113.539601] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The circadian clockworks gate macrophage inflammatory responses. Given the association between clock dysregulation and metabolic disorders, we conducted experiments to determine the extent to which over-nutrition modulates macrophage clock function and whether macrophage circadian dysregulation is a key factor linking over-nutrition to macrophage proinflammatory activation, adipose tissue inflammation, and systemic insulin resistance. Our results demonstrate that 1) macrophages from high fat diet-fed mice are marked by dysregulation of the molecular clockworks in conjunction with increased proinflammatory activation, 2) global disruption of the clock genes Period1 (Per1) and Per2 recapitulates this amplified macrophage proinflammatory activation, 3) adoptive transfer of Per1/2-disrupted bone marrow cells into wild-type mice potentiates high fat diet-induced adipose and liver tissue inflammation and systemic insulin resistance, and 4) Per1/2-disrupted macrophages similarly exacerbate inflammatory responses and decrease insulin sensitivity in co-cultured adipocytes in vitro. Furthermore, PPARγ levels are decreased in Per1/2-disrupted macrophages and PPARγ2 overexpression ameliorates Per1/2 disruption-associated macrophage proinflammatory activation, suggesting that this transcription factor may link the molecular clockworks to signaling pathways regulating macrophage polarization. Thus, macrophage circadian clock dysregulation is a key process in the physiological cascade by which diet-induced obesity triggers macrophage proinflammatory activation, adipose tissue inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Hang Xu
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Honggui Li
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Shih-Lung Woo
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Sam-Moon Kim
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807
| | - Vikram R Shende
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807
| | - Nichole Neuendorff
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807
| | - Xin Guo
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Ting Guo
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Ting Qi
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Ya Pei
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Yan Zhao
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843
| | - Xiang Hu
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843, Department of Endocrinology and
| | - Jiajia Zhao
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843, Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China, and
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China, and
| | | | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine and
| | - Robert C Alaniz
- Department of Microbial and Molecular Pathogenesis, College of Medicine, Texas A&M Health Science Center, College Station, Texas 77843
| | - David J Earnest
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas 77807,
| | - Chaodong Wu
- From the Department of Nutrition and Food Science, Texas A&M University, College Station, Texas 77843,
| |
Collapse
|
46
|
Woo SL, Xu H, Li H, Zhao Y, Hu X, Zhao J, Guo X, Guo T, Botchlett R, Qi T, Pei Y, Zheng J, Xu Y, An X, Chen L, Chen L, Li Q, Xiao X, Huo Y, Wu C. Metformin ameliorates hepatic steatosis and inflammation without altering adipose phenotype in diet-induced obesity. PLoS One 2014; 9:e91111. [PMID: 24638078 PMCID: PMC3956460 DOI: 10.1371/journal.pone.0091111] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 02/06/2014] [Indexed: 01/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely associated with obesity and insulin resistance. To better understand the pathophysiology of obesity-associated NAFLD, the present study examined the involvement of liver and adipose tissues in metformin actions on reducing hepatic steatosis and inflammation during obesity. C57BL/6J mice were fed a high-fat diet (HFD) for 12 weeks to induce obesity-associated NAFLD and treated with metformin (150 mg/kg/d) orally for the last four weeks of HFD feeding. Compared with HFD-fed control mice, metformin-treated mice showed improvement in both glucose tolerance and insulin sensitivity. Also, metformin treatment caused a significant decrease in liver weight, but not adiposity. As indicated by histological changes, metformin treatment decreased hepatic steatosis, but not the size of adipocytes. In addition, metformin treatment caused an increase in the phosphorylation of liver AMP-activated protein kinase (AMPK), which was accompanied by an increase in the phosphorylation of liver acetyl-CoA carboxylase and decreases in the phosphorylation of liver c-Jun N-terminal kinase 1 (JNK1) and in the mRNA levels of lipogenic enzymes and proinflammatory cytokines. However, metformin treatment did not significantly alter adipose tissue AMPK phosphorylation and inflammatory responses. In cultured hepatocytes, metformin treatment increased AMPK phosphorylation and decreased fat deposition and inflammatory responses. Additionally, in bone marrow-derived macrophages, metformin treatment partially blunted the effects of lipopolysaccharide on inducing the phosphorylation of JNK1 and nuclear factor kappa B (NF-κB) p65 and on increasing the mRNA levels of proinflammatory cytokines. Taken together, these results suggest that metformin protects against obesity-associated NAFLD largely through direct effects on decreasing hepatocyte fat deposition and on inhibiting inflammatory responses in both hepatocytes and macrophages.
Collapse
Affiliation(s)
- Shih-Lung Woo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Hang Xu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Yan Zhao
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Xiang Hu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America; Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiajia Zhao
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America; Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Ting Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Rachel Botchlett
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Ting Qi
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| | - Juan Zheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America; Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yiming Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America
| | - Xiaofei An
- Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qifu Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States of America; Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
47
|
Eissing L, Scherer T, Tödter K, Knippschild U, Greve JW, Buurman WA, Pinnschmidt HO, Rensen SS, Wolf AM, Bartelt A, Heeren J, Buettner C, Scheja L. De novo lipogenesis in human fat and liver is linked to ChREBP-β and metabolic health. Nat Commun 2013; 4:1528. [PMID: 23443556 DOI: 10.1038/ncomms2537] [Citation(s) in RCA: 231] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 01/23/2013] [Indexed: 02/07/2023] Open
Abstract
Clinical interest in de novo lipogenesis has been sparked by recent studies in rodents demonstrating that de novo lipogenesis specifically in white adipose tissue produces the insulin-sensitizing fatty acid palmitoleate. By contrast, hepatic lipogenesis is thought to contribute to metabolic disease. How de novo lipogenesis in white adipose tissue versus liver is altered in human obesity and insulin resistance is poorly understood. Here we show that lipogenic enzymes and the glucose transporter-4 are markedly decreased in white adipose tissue of insulin-resistant obese individuals compared with non-obese controls. By contrast, lipogenic enzymes are substantially upregulated in the liver of obese subjects. Bariatric weight loss restored de novo lipogenesis and glucose transporter-4 gene expression in white adipose tissue. Notably, lipogenic gene expression in both white adipose tissue and liver was strongly linked to the expression of carbohydrate-responsive element-binding protein-β and to metabolic risk markers. Thus, de novo lipogenesis predicts metabolic health in humans in a tissue-specific manner and is likely regulated by glucose-dependent carbohydrate-responsive element-binding protein activation.
Collapse
Affiliation(s)
- Leah Eissing
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gly482Ser mutation impairs the effects of peroxisome proliferator-activated receptor γ coactivator-1α on decreasing fat deposition and stimulating phosphoenolpyruvate carboxykinase expression in hepatocytes. Nutr Res 2013; 33:332-9. [PMID: 23602251 DOI: 10.1016/j.nutres.2013.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 12/22/2012] [Accepted: 02/05/2013] [Indexed: 12/21/2022]
Abstract
Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a transcriptional coactivator of nuclear receptor peroxisome proliferator-activated receptor γ that critically regulates glucose and fat metabolism. Although clinical evidence suggests that Gly482Ser polymorphism of PGC-1α is associated with an increased incidence of nonalcoholic fatty liver disease, a direct role for Gly482Ser mutation in altering PGC-1α actions on hepatocyte fat deposition remains to be explored. We hypothesized that Gly482Ser mutation impairs the abilities of PGC-1α in ameliorating overnutrition-induced hepatocyte fat deposition and in stimulating hepatocyte expression of cytosolic phosphoenolpyruvate carboxykinase (PEPCK-C; encoded by a key PGC-1α target gene). In the present study, treatment of cultured hepatocytes with palmitate induced fat deposition, serving as a cell model of hepatic steatosis. Upon overexpression of wild-type PGC-1α, H4IIE cells exhibited a significant decrease in palmitate-induced hepatocyte fat deposition compared with control cells and/or cells upon overexpression of mutant PGC-1α (Gly482Ser). Overexpression of wild-type PGC-1α, but not mutant PGC-1α, also caused a significant increase in hepatocyte expression of carnitine palmitoyl transferase 1a, a rate-determining enzyme that transfers long-chain fatty acids into mitochondria for oxidation. In addition, overexpression of mutant PGC-1α did not stimulate PEPCK-C expression as overexpression of wild-type PGC-1α did, likely due to a decrease in the ability of mutant PGC-1α in increasing PEPCK promoter transcription activity. Together, these results suggest that Gly482Ser mutation impairs the abilities of PGC-1α in decreasing fat deposition and in stimulating PEPCK-C expression in cultured hepatocytes.
Collapse
|
49
|
Li H, Guo X, Xu H, Woo SL, Halim V, Morgan C, Wu C. A role for inducible 6-phosphofructo-2-kinase in the control of neuronal glycolysis. J Nutr Biochem 2012; 24:1153-8. [PMID: 23246158 DOI: 10.1016/j.jnutbio.2012.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 08/07/2012] [Accepted: 08/27/2012] [Indexed: 10/27/2022]
Abstract
Increased glycolysis is the result of the sensing of glucose by hypothalamic neurons. The biochemical mechanisms underlying the control of hypothalamic glycolysis, however, remain to be elucidated. Here we showed that PFKFB3, the gene that encodes for inducible 6-phosphofructo-2-kinase (iPFK2), was expressed at high abundance in both mouse hypothalami and clonal hypothalamic neurons. In response to re-feeding, PFKFB3 mRNA levels were increased by 10-fold in mouse hypothalami. In the hypothalamus, re-feeding also decreased the phosphorylation of AMP-activated protein kinase (AMPK) (Thr172) and the mRNA levels of agouti-related protein (AgRP), and increased the mRNA levels of cocaine-amphetamine-related transcript (CART). Similar results were observed in N-43/5 clonal hypothalamic neurons upon treatment with glucose and/or insulin. In addition, knockdown of PFKFB3/iPFK2 in N-43/5 neurons caused a decrease in rates of glycolysis, which was accompanied by increased AMPK phosphorylation, increased AgRP mRNA levels and decreased CART mRNA levels. In contrast, overexpression of PFKFB3/iPFK2 in N-43/5 neurons caused an increase in glycolysis, which was accompanied by decreased AMPK phosphorylation and decreased AgRP mRNA levels and increased CART mRNA levels. Together, these results suggest that PFKFB3/iPFK2 responds to re-feeding, which in turn stimulates hypothalamic glycolysis and decreases hypothalamic AMPK phosphorylation and alters neuropeptide expression in a pattern that is associated with suppression of food intake.
Collapse
Affiliation(s)
- Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
|