1
|
M JN, Bharadwaj D. The complex web of obesity: from genetics to precision medicine. Expert Rev Endocrinol Metab 2024; 19:403-418. [PMID: 38869356 DOI: 10.1080/17446651.2024.2365785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
INTRODUCTION Obesity is a growing public health concern affecting both children and adults. Since it involves both genetic and environmental components, the management of obesity requires both, an understanding of the underlying genetics and changes in lifestyle. The knowledge of obesity genetics will enable the possibility of precision medicine in anti-obesity medications. AREAS COVERED Here, we explore health complications and the prevalence of obesity. We discuss disruptions in energy balance as a symptom of obesity, examining evolutionary theories, its multi-factorial origins, and heritability. Additionally, we discuss monogenic and polygenic obesity, the converging biological pathways, potential pharmacogenomics applications, and existing anti-obesity medications - specifically focussing on the leptin-melanocortin and incretin pathways. Comparisons between childhood and adult obesity genetics are made, along with insights into structural variants, epigenetic changes, and environmental influences on epigenetic signatures. EXPERT OPINION With recent advancements in anti-obesity drugs, genetic studies pinpoint new targets and allow for repurposing existing drugs. This creates opportunities for genotype-informed treatment options. Also, lifestyle interventions can help in the prevention and treatment of obesity by altering the epigenetic signatures. The comparison of genetic architecture in adults and children revealed a significant overlap. However, more robust studies with diverse ethnic representation is required in childhood obesity.
Collapse
Affiliation(s)
- Janaki Nair M
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Dwaipayan Bharadwaj
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
2
|
Huang X, Chen J, Li H, Cai Y, Liu L, Dong Q, Li Y, Ren Y, Xiang W, He X. LncRNA SNHG12 suppresses adipocyte inflammation and insulin resistance by regulating the HDAC9/Nrf2 axis. FASEB J 2024; 38:e23794. [PMID: 38967258 DOI: 10.1096/fj.202400236rr] [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: 01/30/2024] [Revised: 06/12/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Obesity is often associated with low-grade inflammation. The incidence of obesity has increased annually worldwide, which seriously affects human health. A previous study indicated that long noncoding RNA SNHG12 was downregulated in obesity. Nevertheless, the role of SNHG12 in obesity remains to be elucidated. In this study, qRT-PCR, western blot, and ELISA were utilized to examine the gene and protein expression. Flow cytometry was employed to investigate the M2 macrophage markers. RNA pull-down assay and RIP were utilized to confirm the interactions of SNHG12, hnRNPA1, and HDAC9. Eventually, a high-fat diet-fed mouse model was established for in vivo studies. SNHG12 overexpression suppressed adipocyte inflammation and insulin resistance and promoted M2 polarization of macrophages that was caused by TNF-α treatment. SNHG12 interacted with hnRNPA1 to downregulate HDAC9 expression, which activated the Nrf2 signaling pathway. HDAC9 overexpression reversed the effect of SNHG12 overexpression on inflammatory response, insulin resistance, and M2 phenotype polarization. Overexpression of SNHG12 improved high-fat diet-fed mouse tissue inflammation. This study revealed the protective effect of SNHG12 against adipocyte inflammation and insulin resistance. This result further provides a new therapeutic target for preventing inflammation and insulin resistance in obesity.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Department of Genetics, Metabolism and Endocrinology, Hainan Women and Children's Medical Center, Haikou, China
| | - Jixiong Chen
- Department of Medical Care Center, Hainan Provincial People's Hospital, Haikou, China
| | - Haidan Li
- Department of Genetics, Metabolism and Endocrinology, Hainan Women and Children's Medical Center, Haikou, China
| | - Yuhua Cai
- Department of Genetics, Metabolism and Endocrinology, Hainan Women and Children's Medical Center, Haikou, China
| | - Li Liu
- Department of Genetics, Metabolism and Endocrinology, Hainan Women and Children's Medical Center, Haikou, China
| | - Qi Dong
- Department of Genetics, Metabolism and Endocrinology, Hainan Women and Children's Medical Center, Haikou, China
| | - Yan Li
- Department of Genetics, Metabolism and Endocrinology, Hainan Women and Children's Medical Center, Haikou, China
| | - Yi Ren
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou, China
| | - Wei Xiang
- Hainan Women and Children's Medical Center, Haikou, China
| | - Xiaojie He
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Dang Z, Li H, Xue S, Shao B, Ning Y, Su G, Zhang F, Yu W, Leng S. Histone deacetylase 9-mediated phenotypic transformation of vascular smooth muscle cells is a potential target for treating aortic aneurysm/dissection. Biomed Pharmacother 2024; 173:116396. [PMID: 38460370 DOI: 10.1016/j.biopha.2024.116396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
Aortic aneurysm/dissection (AAD) is a serious cardiovascular condition characterized by rapid onset and high mortality rates. Currently, no effective drug treatment options are known for AAD. AAD pathogenesis is associated with the phenotypic transformation and abnormal proliferation of vascular smooth muscle cells (VSMCs). However, endogenous factors that contribute to AAD progression remain unclear. We aimed to investigate the role of histone deacetylase 9 (HDAC9) in AAD pathogenesis. HDAC9 expression was considerably increased in human thoracic aortic dissection specimens. Using RNA-sequencing (RNA-seq) and chromatin immunoprecipitation, we demonstrated that HDAC9 transcriptionally inhibited the expression of superoxide dismutase 2 and insulin-like growth factor-binding protein-3, which are critically involved in various signaling pathways. Furthermore, HDAC9 triggered the transformation of VSMCs from a systolic to synthetic phenotype, increasing their proliferation and migration abilities and suppressing their apoptosis. Consistent with these results, in vivo experiments revealed that TMP195, a pharmacological inhibitor of HDAC9, suppressed the formation of the β-aminopropionitrile-induced AAD phenotype in mice. Our findings indicate that HDAC9 may be a novel endogenous risk factor that promotes the onset of AAD by mediating the phenotypic transformation of VSMCs. Therefore, HDAC9 may serve as a potential therapeutic target for drug-based AAD treatment. Furthermore, TMP195 holds potential as a therapeutic agent for AAD treatment.
Collapse
Affiliation(s)
- Zhiqiao Dang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Haijie Li
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Shishan Xue
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Baowei Shao
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Yansong Ning
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Guohai Su
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Fengquan Zhang
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China.
| | - Wenqian Yu
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China.
| | - Shuai Leng
- Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China.
| |
Collapse
|
4
|
Byeon HE, Choi SE, Kim Y, Choi S, Lee SJ, Kim DH, Mo JS, Jeon JY. HDAC11 Regulates Palmitate-induced NLRP3 Inflammasome Activation by Inducing YAP Expression in THP-1 Cells and PBMCs. Endocrinology 2024; 165:bqae011. [PMID: 38366363 DOI: 10.1210/endocr/bqae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Indexed: 02/18/2024]
Abstract
Histone deacetylase 11 (HDAC11) has been implicated in the pathogenesis of metabolic diseases characterized by chronic low-grade inflammation, such as obesity. However, the influence of HDAC11 on inflammation and the specific effect of HDAC11 on the palmitic acid (PA)-induced NLR family pyrin domain containing 3 (NLRP3) inflammasome activation are poorly understood. The effect of PA treatment on HDAC11 activity and the NLRP3 inflammasome was investigated in human peripheral blood mononuclear cells and THP-1 cells. The PA-induced responses of key markers of NLRP3 inflammasome activation, including NLRP3 gene expression, caspase-1 p10 activation, cleaved IL-1β production, and extracellular IL-1β release, were assessed as well. The role of HDAC11 was explored using a specific inhibitor of HDAC11 and by knockdown using small interfering (si)HDAC11 RNA. The relationship between HDAC11 and yes-associated protein (YAP) in the PA-induced NLRP3 inflammasome was investigated in THP-1 cells with HDAC11 or YAP knockdown. Following PA treatment, HDAC11 activity and protein levels increased significantly, concomitant with activation of the NLRP3 inflammasome. Notably, PA-induced the upregulation of NLRP3, caspase-1 p10 activation, the production of cleaved IL-1β, and the release of IL-1β into the extracellular space, all of which were attenuated by FT895 treatment and by HDAC11 knockdown. In THP-1 cells, PA induced the expression of YAP and its interaction with NLRP3, resulting in NLRP3 inflammasome activation, whereas both were inhibited by FT895 and siHDAC11 RNA. These findings demonstrate a pivotal role for HDAC11 in the PA-induced activation of the NLRP3 inflammasome. HDAC11 inhibition thus represents a promising therapeutic strategy for mitigating NLRP3 inflammasome-related inflammation in the context of obesity.
Collapse
Affiliation(s)
- Hye-Eun Byeon
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Sung-E Choi
- Department of Physiology, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Yujin Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
- Department of Biomedical Sciences, Graduate School of Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Suji Choi
- Department of Biological Sciences, Hyupsung University, Hwasung-si, Gyeonggi-do 18330, Republic of Korea
| | - Soo-Jin Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Dong Hyun Kim
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
- Department of Biomedical Sciences, Graduate School of Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Jung-Soon Mo
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| | - Ja Young Jeon
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Gyeonggi-do 16499, Republic of Korea
| |
Collapse
|
5
|
Das T, Khatun S, Jha T, Gayen S. HDAC9 as a Privileged Target: Reviewing its Role in Different Diseases and Structure-activity Relationships (SARs) of its Inhibitors. Mini Rev Med Chem 2024; 24:767-784. [PMID: 37818566 DOI: 10.2174/0113895575267301230919165827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/17/2023] [Accepted: 08/11/2023] [Indexed: 10/12/2023]
Abstract
HDAC9 is a histone deacetylase enzyme belonging to the class IIa of HDACs which catalyses histone deacetylation. HDAC9 inhibit cell proliferation by repairing DNA, arresting the cell cycle, inducing apoptosis, and altering genetic expression. HDAC9 plays a significant part in human physiological system and are involved in various type of diseases like cancer, diabetes, atherosclerosis and CVD, autoimmune response, inflammatory disease, osteoporosis and liver fibrosis. This review discusses the role of HDAC9 in different diseases and structure-activity relationships (SARs) of various hydroxamate and non-hydroxamate-based inhibitors. SAR of compounds containing several scaffolds have been discussed in detail. Moreover, structural requirements regarding the various components of HDAC9 inhibitor (cap group, linker and zinc-binding group) has been highlighted in this review. Though, HDAC9 is a promising target for the treatment of a number of diseases including cancer, a very few research are available. Thus, this review may provide useful information for designing novel HDAC9 inhibitors to fight against different diseases in the future.
Collapse
Affiliation(s)
- Totan Das
- Department of Pharmaceutical Technology, Laboratory of Drug Design and Discovery, Jadavpur University, Kolkata, 700032, India
| | - Samima Khatun
- Department of Pharmaceutical Technology, Laboratory of Drug Design and Discovery, Jadavpur University, Kolkata, 700032, India
| | - Tarun Jha
- Department of Pharmaceutical Technology, Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Jadavpur University, Kolkata, 700032, India
| | - Shovanlal Gayen
- Department of Pharmaceutical Technology, Laboratory of Drug Design and Discovery, Jadavpur University, Kolkata, 700032, India
| |
Collapse
|
6
|
Machado FR, Boeira SP, Bortolotto VC, Araujo SM, Poetini MR, Viana CE, Prigol M, Souza LC, de Gomes MG. HDAC3 inhibition protects against peripheral and central alterations in an animal model of obesity. Pharmacol Rep 2023; 75:1177-1186. [PMID: 37698830 DOI: 10.1007/s43440-023-00528-7] [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: 06/22/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND Obesity is a multifactorial disease with epigenetic manifestations that increases the prevalence of associated comorbidities such as metabolic syndrome, cardiovascular dysfunction, and major depression disorder. Given the aforementioned, a search for new pharmacological alternatives for the treatment of this disease is necessary. The current study aimed to evaluate the effects of histone deacetylase-3 (HDAC3) inhibition caused by RGFP966 (a benzamide-type HDAC inhibitor selective for HDAC3) administration, in an animal model of obesity induced by high-fat diet (HFD). METHODS Adult male mice C57BJ/6 were fed with a normal pellet diet (NPD) or HFD for 120 days. The HDAC3 inhibitor (RGFP966; 10 mg/kg; sc) was administered on the 91st to 120th day of the experiment (per 30 days). After the last inhibitor administration, animals were euthanized, blood was collected, and the hippocampus was removed for biochemical determinations. RESULTS In an overall manner, the administration of RGFP966 protected against changes in body weight gain, glucose, insulin, lipid profile, adipokines, and increase of hippocampal proinflammatory cytokines levels caused by HFD. CONCLUSION Therefore, HDAC3 inhibition can represent a promising pharmacological target for the treatment of obesity.
Collapse
Affiliation(s)
- Franciéle Romero Machado
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Silvana Peterini Boeira
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Vandreza Cardoso Bortolotto
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Stífani Machado Araujo
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Márcia Rósula Poetini
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Cristini Escobar Viana
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Marina Prigol
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Leandro Cattelan Souza
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil
| | - Marcelo Gomes de Gomes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, LaftamBio Pampa, Federal University of Pampa, Campus Itaqui, Itaqui, RS, 97650-000, Brazil.
| |
Collapse
|
7
|
Soukar I, Amarasinghe A, Pile LA. Coordination of cross-talk between metabolism and epigenetic regulation by the SIN3 complex. Enzymes 2023; 53:33-68. [PMID: 37748836 DOI: 10.1016/bs.enz.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Post-translational modifications of histone proteins control the expression of genes. Metabolites from central and one-carbon metabolism act as donor moieties to modify histones and regulate gene expression. Thus, histone modification and gene regulation are connected to the metabolite status of the cell. Histone modifiers, such as the SIN3 complex, regulate genes involved in proliferation and metabolism. The SIN3 complex contains a histone deacetylase and a histone demethylase, which regulate the chromatin landscape and gene expression. In this chapter, we review the cross-talk between metabolic pathways that produce donor moieties, and epigenetic complexes regulating proliferation and metabolic genes. This cross-talk between gene regulation and metabolism is tightly controlled, and disruption of this cross-talk leads to metabolic diseases. We discuss promising therapeutics that directly regulate histone modifiers, and can affect the metabolic status of the cell, alleviating some metabolic diseases.
Collapse
Affiliation(s)
- Imad Soukar
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Anjalie Amarasinghe
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Lori A Pile
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States.
| |
Collapse
|
8
|
Zhou R, Cao Y, Xiang Y, Fang P, Shang W. Emerging roles of histone deacetylases in adaptive thermogenesis. Front Endocrinol (Lausanne) 2023; 14:1124408. [PMID: 36875455 PMCID: PMC9978507 DOI: 10.3389/fendo.2023.1124408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Brown and beige adipose tissues regulate body energy expenditure through adaptive thermogenesis, which converts energy into heat by oxidative phosphorylation uncoupling. Although promoting adaptive thermogenesis has been demonstrated to be a prospective strategy for obesity control, there are few methods for increasing adipose tissue thermogenesis in a safe and effective way. Histone deacetylase (HDAC) is a category of epigenetic modifying enzymes that catalyzes deacetylation on both histone and non-histone proteins. Recent studies illustrated that HDACs play an important role in adipose tissue thermogenesis through modulating gene transcription and chromatin structure as well as cellular signals transduction in both deacetylation dependent or independent manners. Given that different classes and subtypes of HDACs show diversity in the mechanisms of adaptive thermogenesis regulation, we systematically summarized the effects of different HDACs on adaptive thermogenesis and their underlying mechanisms in this review. We also emphasized the differences among HDACs in thermogenesis regulation, which will help to find new efficient anti-obesity drugs targeting specific HDAC subtypes.
Collapse
Affiliation(s)
- Ruonan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Cao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yingying Xiang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Penghua Fang, ; Wenbin Shang,
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Penghua Fang, ; Wenbin Shang,
| |
Collapse
|
9
|
Mahmoud AM. An Overview of Epigenetics in Obesity: The Role of Lifestyle and Therapeutic Interventions. Int J Mol Sci 2022; 23:ijms23031341. [PMID: 35163268 PMCID: PMC8836029 DOI: 10.3390/ijms23031341] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity has become a global epidemic that has a negative impact on population health and the economy of nations. Genetic predispositions have been demonstrated to have a substantial role in the unbalanced energy metabolism seen in obesity. However, these genetic variations cannot entirely explain the massive growth in obesity over the last few decades. Accumulating evidence suggests that modern lifestyle characteristics such as the intake of energy-dense foods, adopting sedentary behavior, or exposure to environmental factors such as industrial endocrine disruptors all contribute to the rising obesity epidemic. Recent advances in the study of DNA and its alterations have considerably increased our understanding of the function of epigenetics in regulating energy metabolism and expenditure in obesity and metabolic diseases. These epigenetic modifications influence how DNA is transcribed without altering its sequence. They are dynamic, reflecting the interplay between the body and its surroundings. Notably, these epigenetic changes are reversible, making them appealing targets for therapeutic and corrective interventions. In this review, I discuss how these epigenetic modifications contribute to the disordered energy metabolism in obesity and to what degree lifestyle and weight reduction strategies and pharmacological drugs can restore energy balance by restoring normal epigenetic profiles.
Collapse
Affiliation(s)
- Abeer M Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
10
|
Lecce L, Xu Y, V’Gangula B, Chandel N, Pothula V, Caudrillier A, Santini MP, d’Escamard V, Ceholski DK, Gorski PA, Ma L, Koplev S, Bjørklund MM, Björkegren JL, Boehm M, Bentzon JF, Fuster V, Kim HW, Weintraub NL, Baker AH, Bernstein E, Kovacic JC. Histone deacetylase 9 promotes endothelial-mesenchymal transition and an unfavorable atherosclerotic plaque phenotype. J Clin Invest 2021; 131:131178. [PMID: 34338228 PMCID: PMC8321575 DOI: 10.1172/jci131178] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 06/11/2021] [Indexed: 01/05/2023] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is associated with various cardiovascular diseases and in particular with atherosclerosis and plaque instability. However, the molecular pathways that govern EndMT are poorly defined. Specifically, the role of epigenetic factors and histone deacetylases (HDACs) in controlling EndMT and the atherosclerotic plaque phenotype remains unclear. Here, we identified histone deacetylation, specifically that mediated by HDAC9 (a class IIa HDAC), as playing an important role in both EndMT and atherosclerosis. Using in vitro models, we found class IIa HDAC inhibition sustained the expression of endothelial proteins and mitigated the increase in mesenchymal proteins, effectively blocking EndMT. Similarly, ex vivo genetic knockout of Hdac9 in endothelial cells prevented EndMT and preserved a more endothelial-like phenotype. In vivo, atherosclerosis-prone mice with endothelial-specific Hdac9 knockout showed reduced EndMT and significantly reduced plaque area. Furthermore, these mice displayed a more favorable plaque phenotype, with reduced plaque lipid content and increased fibrous cap thickness. Together, these findings indicate that HDAC9 contributes to vascular pathology by promoting EndMT. Our study provides evidence for a pathological link among EndMT, HDAC9, and atherosclerosis and suggests that targeting of HDAC9 may be beneficial for plaque stabilization or slowing the progression of atherosclerotic disease.
Collapse
Affiliation(s)
- Laura Lecce
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yang Xu
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bhargavi V’Gangula
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nirupama Chandel
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Venu Pothula
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Axelle Caudrillier
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Maria Paola Santini
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Valentina d’Escamard
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Delaine K. Ceholski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Przemek A. Gorski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lijiang Ma
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Simon Koplev
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Martin Mæng Bjørklund
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Johan L.M. Björkegren
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Jacob Fog Bentzon
- Department of Clinical Medicine, Heart Diseases, Aarhus University, Aarhus, Denmark
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Valentin Fuster
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ha Won Kim
- Department of Medicine, Cardiology Division and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Neal L. Weintraub
- Department of Medicine, Cardiology Division and Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Andrew H. Baker
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Emily Bernstein
- Departments of Oncological Sciences and Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jason C. Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- St. Vincent’s Clinical School, University of New South Wales, Sydney, Australia
| |
Collapse
|
11
|
Asif S, Morrow NM, Mulvihill EE, Kim KH. Understanding Dietary Intervention-Mediated Epigenetic Modifications in Metabolic Diseases. Front Genet 2020; 11:590369. [PMID: 33193730 PMCID: PMC7593700 DOI: 10.3389/fgene.2020.590369] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
The global prevalence of metabolic disorders, such as obesity, diabetes and fatty liver disease, is dramatically increasing. Both genetic and environmental factors are well-known contributors to the development of these diseases and therefore, the study of epigenetics can provide additional mechanistic insight. Dietary interventions, including caloric restriction, intermittent fasting or time-restricted feeding, have shown promising improvements in patients' overall metabolic profiles (i.e., reduced body weight, improved glucose homeostasis), and an increasing number of studies have associated these beneficial effects with epigenetic alterations. In this article, we review epigenetic changes involved in both metabolic diseases and dietary interventions in primary metabolic tissues (i.e., adipose, liver, and pancreas) in hopes of elucidating potential biomarkers and therapeutic targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Shaza Asif
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Nadya M. Morrow
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Erin E. Mulvihill
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Kyoung-Han Kim
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
12
|
Asare Y, Campbell-James TA, Bokov Y, Yu LL, Prestel M, El Bounkari O, Roth S, Megens RTA, Straub T, Thomas K, Yan G, Schneider M, Ziesch N, Tiedt S, Silvestre-Roig C, Braster Q, Huang Y, Schneider M, Malik R, Haffner C, Liesz A, Soehnlein O, Bernhagen J, Dichgans M. Histone Deacetylase 9 Activates IKK to Regulate Atherosclerotic Plaque Vulnerability. Circ Res 2020; 127:811-823. [PMID: 32546048 DOI: 10.1161/circresaha.120.316743] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Arterial inflammation manifested as atherosclerosis is the leading cause of mortality worldwide. Genome-wide association studies have identified a prominent role of HDAC (histone deacetylase)-9 in atherosclerosis and its clinical complications including stroke and myocardial infarction. OBJECTIVE To determine the mechanisms linking HDAC9 to these vascular pathologies and explore its therapeutic potential for atheroprotection. METHODS AND RESULTS We studied the effects of Hdac9 on features of plaque vulnerability using bone marrow reconstitution experiments and pharmacological targeting with a small molecule inhibitor in hyperlipidemic mice. We further used 2-photon and intravital microscopy to study endothelial activation and leukocyte-endothelial interactions. We show that hematopoietic Hdac9 deficiency reduces lesional macrophage content while increasing fibrous cap thickness thus conferring plaque stability. We demonstrate that HDAC9 binds to IKK (inhibitory kappa B kinase)-α and β, resulting in their deacetylation and subsequent activation, which drives inflammatory responses in both macrophages and endothelial cells. Pharmacological inhibition of HDAC9 with the class IIa HDAC inhibitor TMP195 attenuates lesion formation by reducing endothelial activation and leukocyte recruitment along with limiting proinflammatory responses in macrophages. Transcriptional profiling using RNA sequencing revealed that TMP195 downregulates key inflammatory pathways consistent with inhibitory effects on IKKβ. TMP195 mitigates the progression of established lesions and inhibits the infiltration of inflammatory cells. Moreover, TMP195 diminishes features of plaque vulnerability and thereby enhances plaque stability in advanced lesions. Ex vivo treatment of monocytes from patients with established atherosclerosis reduced the production of inflammatory cytokines including IL (interleukin)-1β and IL-6. CONCLUSIONS Our findings identify HDAC9 as a regulator of atherosclerotic plaque stability and IKK activation thus providing a mechanistic explanation for the prominence of HDAC9 as a vascular risk locus in genome-wide association studies. Its therapeutic inhibition may provide a potent lever to alleviate vascular inflammation. Graphical Abstract: A graphical abstract is available for this article.
Collapse
Affiliation(s)
- Yaw Asare
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Thomas A Campbell-James
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Yury Bokov
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Lydia Luya Yu
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Matthias Prestel
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Omar El Bounkari
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Roth
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention (R.T.A.M., C.S.-R., Q.B., O.S.), Ludwig-Maximilians-University, Munich, Germany.,Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (R.T.A.M.)
| | - Tobias Straub
- BMC, Core Facility Bioinformatics Munich, Germany (T.S.)
| | - Kyra Thomas
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Guangyao Yan
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Melanie Schneider
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Natalie Ziesch
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Steffen Tiedt
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Carlos Silvestre-Roig
- Institute for Cardiovascular Prevention (R.T.A.M., C.S.-R., Q.B., O.S.), Ludwig-Maximilians-University, Munich, Germany
| | - Quinte Braster
- Institute for Cardiovascular Prevention (R.T.A.M., C.S.-R., Q.B., O.S.), Ludwig-Maximilians-University, Munich, Germany
| | - Yishu Huang
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Manuela Schneider
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Rainer Malik
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Christof Haffner
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany
| | - Arthur Liesz
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology, Germany (A.L., J.B., M.D.)
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (R.T.A.M., C.S.-R., Q.B., O.S.), Ludwig-Maximilians-University, Munich, Germany.,German Center for Cardiovascular Research, Partner Site Munich Heart Alliance (O.S., J.B.).,Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (O.S.)
| | - Jürgen Bernhagen
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology, Germany (A.L., J.B., M.D.).,German Center for Cardiovascular Research, Partner Site Munich Heart Alliance (O.S., J.B.)
| | - Martin Dichgans
- From the Institute for Stroke and Dementia Research, University Hospital (Y.A., T.A.C.-J., Y.B., L.L.Y., M.P., O.E.B., S.R., K.T., G.Y., M.S., N.Z., S.T., Y.H., M.S., R.M., C.H., A.L., J.B., M.D.), Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology, Germany (A.L., J.B., M.D.)
| |
Collapse
|
13
|
Khamis A, Boutry R, Canouil M, Mathew S, Lobbens S, Crouch H, Andrew T, Abderrahmani A, Tamanini F, Froguel P. Histone deacetylase 9 promoter hypomethylation associated with adipocyte dysfunction is a statin-related metabolic effect. Clin Epigenetics 2020; 12:68. [PMID: 32410704 PMCID: PMC7222462 DOI: 10.1186/s13148-020-00858-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Background Adipogenesis, the process whereby preadipocytes differentiate into mature adipocytes, is crucial for maintaining metabolic homeostasis. Cholesterol-lowering statins increase type 2 diabetes (T2D) risk possibly by affecting adipogenesis and insulin resistance but the (epi)genetic mechanisms involved are unknown. Here, we characterised the effects of statin treatment on adipocyte differentiation using in vitro human preadipocyte cell model to identify putative effective genes. Results Statin treatment during adipocyte differentiation caused a reduction in key genes involved in adipogenesis, such as ADIPOQ, GLUT4 and ABCG1. Using Illumina’s Infinium ‘850K’ Methylation EPIC array, we found a significant hypomethylation of cg14566882, located in the promoter of the histone deacetylase 9 (HDAC9) gene, in response to two types of statins (atorvastatin and mevastatin), which correlates with an increased HDAC9 mRNA expression. We confirmed that HDAC9 is a transcriptional repressor of the cholesterol efflux ABCG1 gene expression, which is epigenetically modified in obesity and prediabetic states. Thus, we assessed the putative impact of ABCG1 knockdown in mimicking the effect of statin in adipogenesis. ABCG1 KD reduced the expression of key genes involved in adipocyte differentiation and decreased insulin signalling and glucose uptake. In human blood cells from two cohorts, ABCG1 expression was impaired in response to statins, confirming that ABCG1 is targeted in vivo by these drugs. Conclusions We identified an epigenetic link between adipogenesis and adipose tissue insulin resistance in the context of T2D risk associated with statin use, which has important implications as HDAC9 and ABCG1 are considered potential therapeutic targets for obesity and metabolic diseases.
Collapse
Affiliation(s)
- Amna Khamis
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,Université de Lille, Inserm UMR1283, CNRS-UMR 8199 - EGID, Lille, Lille University Hospital, F-59000, Lille, France
| | - Raphael Boutry
- Université de Lille, Inserm UMR1283, CNRS-UMR 8199 - EGID, Lille, Lille University Hospital, F-59000, Lille, France
| | - Mickaël Canouil
- Université de Lille, Inserm UMR1283, CNRS-UMR 8199 - EGID, Lille, Lille University Hospital, F-59000, Lille, France
| | - Sumi Mathew
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stephane Lobbens
- Université de Lille, Inserm UMR1283, CNRS-UMR 8199 - EGID, Lille, Lille University Hospital, F-59000, Lille, France
| | - Hutokshi Crouch
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Toby Andrew
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Amar Abderrahmani
- Université de Lille, Inserm UMR1283, CNRS-UMR 8199 - EGID, Lille, Lille University Hospital, F-59000, Lille, France
| | - Filippo Tamanini
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Philippe Froguel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK. .,Université de Lille, Inserm UMR1283, CNRS-UMR 8199 - EGID, Lille, Lille University Hospital, F-59000, Lille, France.
| |
Collapse
|
14
|
Jannat Ali Pour N, Meshkani R, Toolabi K, Mohassel Azadi S, Zand S, Emamgholipour S. Adipose tissue mRNA expression of HDAC1, HDAC3 and HDAC9 in obese women in relation to obesity indices and insulin resistance. Mol Biol Rep 2020; 47:3459-3468. [PMID: 32277440 DOI: 10.1007/s11033-020-05431-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022]
Abstract
It is well-established that an impaired adipose tissue function and morphology caused by a dysregulated gene expression contribute substantially to obesity. Nowadays, animal model studies and in vitro surveys provide evidence for possible roles of HDACs as emerging epigenetic players in the pathogenesis of obesity. However, the clinical pertinence of HDACs in the field of obesity research in humans is not yet obvious. Here, we investigated mRNA expression of HDAC1, 3 and 9 in visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) of obese female participants (n = 20) and normal-weight women (n = 19). We also evaluated the association of the afore-mentioned HDACs gene expression with obesity indices, insulin resistance parameters, and other obesity-related characteristics. Our data revealed the mRNA level of HDAC1 was significantly decreased in both VAT and SAT of obese women, compared to controls. Moreover, the SAT mRNA expression of HDAC3 and VAT mRNA levels of HDAC9 were significantly lower in obese subjects than those found in controls. We observed that HDAC1 and HDAC3 expression in adipose tissue from the whole population is inversely correlated with obesity indices; BMI, waist, hip and waist-to-height ratio (WHtR). Moreover, we found that HDAC3 expression in adipose tissue had an inverse correlation with HOMA-IR, insulin levels, and serum concentration of hs-CRP. Moreover, VAT HDAC9 mRNA level is inversely correlated with obesity indices; BMI, waist, hip and WHtR and with HOMA-IR, insulin levels, and serum concentration of hs-CRP. Hence, it seems that decreased HDAC1,3 and 9 mRNA expression in adipose tissue might be associated with obesity and related abnormalities. However, more studies are needed to establish this concept.
Collapse
Affiliation(s)
- Naghmeh Jannat Ali Pour
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Karamollah Toolabi
- Department of Surgery, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Mohassel Azadi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahabedin Zand
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solaleh Emamgholipour
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Hu S, Cho EH, Lee JY. Histone Deacetylase 9: Its Role in the Pathogenesis of Diabetes and Other Chronic Diseases. Diabetes Metab J 2020; 44:234-244. [PMID: 32347025 PMCID: PMC7188980 DOI: 10.4093/dmj.2019.0243] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 12/11/2022] Open
Abstract
As a member of the class IIa histone deacetylases (HDACs), HDAC9 catalyzes the deacetylation of histones and transcription factors, commonly leading to the suppression of gene transcription. The activity of HDAC9 is regulated transcriptionally and post-translationally. HDAC9 is known to play an essential role in regulating myocyte and adipocyte differentiation and cardiac muscle development. Also, recent studies have suggested that HDAC9 is involved in the pathogenesis of chronic diseases, including cardiovascular diseases, osteoporosis, autoimmune disease, cancer, obesity, insulin resistance, and liver fibrosis. HDAC9 modulates the expression of genes related to the pathogenesis of chronic diseases by altering chromatin structure in their promotor region or reducing the transcriptional activity of their respective transcription factors. This review summarizes the current knowledge of the regulation of HDAC9 expression and activity. Also, the roles of HDAC9 in the pathogenesis of chronic diseases are discussed, along with potential underlying mechanisms.
Collapse
Affiliation(s)
- Siqi Hu
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA
| | - Eun Hee Cho
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Ji Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
16
|
Yi D, Nguyen HP, Sul HS. Epigenetic dynamics of the thermogenic gene program of adipocytes. Biochem J 2020; 477:1137-1148. [PMID: 32219439 PMCID: PMC8594062 DOI: 10.1042/bcj20190599] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
Brown adipose tissue (BAT) is a metabolically beneficial organ capable of burning fat by dissipating chemical energy into heat, thereby increasing energy expenditure. Moreover, subcutaneous white adipose tissue can undergo so-called browning/beiging. The recent recognition of the presence of brown or beige adipocytes in human adults has attracted much attention to elucidate the molecular mechanism underlying the thermogenic adipose program. Many key transcriptional regulators critical for the thermogenic gene program centering on activating the UCP1 promoter, have been discovered. Thermogenic gene expression in brown adipocytes rely on co-ordinated actions of a multitude of transcription factors, including EBF2, PPARγ, Zfp516 and Zc3h10. These transcription factors probably integrate into a cohesive network for BAT gene program. Moreover, these transcription factors recruit epigenetic factors, such as LSD1 and MLL3/4, for specific histone signatures to establish the favorable chromatin landscape. In this review, we discuss advances made in understanding the molecular mechanism underlying the thermogenic gene program, particularly epigenetic regulation.
Collapse
Affiliation(s)
- Danielle Yi
- Department of Nutritional Sciences and Toxicology and Endocrinology Program, University of California, Berkeley, CA 94720, U.S.A
| | - Hai P Nguyen
- Department of Nutritional Sciences and Toxicology and Endocrinology Program, University of California, Berkeley, CA 94720, U.S.A
| | - Hei Sook Sul
- Department of Nutritional Sciences and Toxicology and Endocrinology Program, University of California, Berkeley, CA 94720, U.S.A
| |
Collapse
|
17
|
Concise Review: The Regulatory Mechanism of Lysine Acetylation in Mesenchymal Stem Cell Differentiation. Stem Cells Int 2020; 2020:7618506. [PMID: 32399051 PMCID: PMC7204305 DOI: 10.1155/2020/7618506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/02/2020] [Indexed: 12/30/2022] Open
Abstract
Nowadays, the use of MSCs has attracted considerable attention in the global science and technology field, with the self-renewal and multidirectional differentiation potential for diabetes, obesity treatment, bone repair, nerve repair, myocardial repair, and so on. Epigenetics plays an important role in the regulation of mesenchymal stem cell differentiation, which has become a research hotspot in the medical field. This review focuses on the role of lysine acetylation modification on the determination of MSC differentiation direction. During this progress, the recruitment of lysine acetyltransferases (KATs) and lysine deacetylases (KDACs) is the crux of transcriptional mechanisms in the dynamic regulation of key genes controlling MSC multidirectional differentiation.
Collapse
|
18
|
Patsouras M, Karagianni P, Kogionou P, Vlachoyiannopoulos P. Differential CpG methylation of the promoter of interleukin 8 and the first intron of tissue factor in Antiphospholipid syndrome. J Autoimmun 2019; 102:159-166. [DOI: 10.1016/j.jaut.2019.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/01/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
|
19
|
Abstract
The dramatic increase in global prevalence of metabolic disease is inexplicable when considering only environmental or only genetic factors, leading to the need to explore the possible roles of epigenetic factors. A great deal of progress has been made in this interdisciplinary field in recent years, with many studies investigating various aspects of the metabolic syndrome and its associated epigenetic changes. Rodent models of metabolic diseases have been particularly illuminating because of the ability to leverage tools such as genetic and environmental modifications. The current review summarizes recent breakthroughs regarding epigenetic markers in studies of obesity, Type II diabetes, and cardiovascular disease, the three major disorders associated with metabolic syndrome. We also discuss open questions and future directions for integrating genomic, epigenomic, and phenotypic big biodata toward understanding metabolic syndrome etiology.
Collapse
Affiliation(s)
- Caryn Carson
- Department of Genetics, Washington University School of Medicine , Saint Louis, Missouri
| | - Heather A Lawson
- Department of Genetics, Washington University School of Medicine , Saint Louis, Missouri
| |
Collapse
|
20
|
Bagchi RA, Ferguson BS, Stratton MS, Hu T, Cavasin MA, Sun L, Lin YH, Liu D, Londono P, Song K, Pino MF, Sparks LM, Smith SR, Scherer PE, Collins S, Seto E, McKinsey TA. HDAC11 suppresses the thermogenic program of adipose tissue via BRD2. JCI Insight 2018; 3:120159. [PMID: 30089714 PMCID: PMC6129125 DOI: 10.1172/jci.insight.120159] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
Little is known about the biological function of histone deacetylase 11 (HDAC11), which is the lone class IV HDAC. Here, we demonstrate that deletion of HDAC11 in mice stimulates brown adipose tissue (BAT) formation and beiging of white adipose tissue (WAT). Consequently, HDAC11-deficient mice exhibit enhanced thermogenic potential and, in response to high-fat feeding, attenuated obesity, improved insulin sensitivity, and reduced hepatic steatosis. Ex vivo and cell-based assays revealed that HDAC11 catalytic activity suppresses the BAT transcriptional program, in both the basal state and in response to β-adrenergic receptor signaling, through a mechanism that is dependent on physical association with BRD2, a bromodomain and extraterminal (BET) acetyl-histone-binding protein. These findings define an epigenetic pathway for the regulation of energy homeostasis and suggest the potential for HDAC11-selective inhibitors for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Rushita A. Bagchi
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Matthew S. Stratton
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tianjing Hu
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria A. Cavasin
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lei Sun
- George Washington University Cancer Center, Washington, DC, USA
| | - Ying-Hsi Lin
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Dianxin Liu
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Pilar Londono
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kunhua Song
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria F. Pino
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida, USA
| | - Lauren M. Sparks
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida, USA
| | - Steven R. Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sheila Collins
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Edward Seto
- George Washington University Cancer Center, Washington, DC, USA
| | - Timothy A. McKinsey
- Department of Medicine, Division of Cardiology, and
- Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
21
|
Sun L, Marin de Evsikova C, Bian K, Achille A, Telles E, Pei H, Seto E. Programming and Regulation of Metabolic Homeostasis by HDAC11. EBioMedicine 2018; 33:157-168. [PMID: 29958910 PMCID: PMC6085537 DOI: 10.1016/j.ebiom.2018.06.025] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/22/2022] Open
Abstract
Histone deacetylases (HDACs) are enzymes that regulate protein functions by catalyzing the removal of acetyl and acyl groups from lysine residues. They play pivotal roles in governing cell behaviors and are indispensable in numerous biological processes. HDAC11, the last identified and sole member of class IV HDACs, was reported over a decade ago. However, its physiological function remains poorly understood. Here, we report that HDAC11 knockout mice are resistant to high-fat diet-induced obesity and metabolic syndrome, suggesting that HDAC11 functions as a crucial metabolic regulator. Depletion of HDAC11 significantly enhanced insulin sensitivity and glucose tolerance, attenuated hypercholesterolemia, and decreased hepatosteatosis and liver damage. Mechanistically, HDAC11 deficiency boosts energy expenditure through promoting thermogenic capacity, which attributes to the elevation of uncoupling protein 1 (UCP1) expression and activity in brown adipose tissue. Moreover, loss of HDAC11 activates the adiponectin-AdipoR-AMPK pathway in the liver, which may contribute to a reversal in hepatosteatosis. Overall, our findings distinguish HDAC11 as a novel regulator of obesity, with potentially important implications for obesity-related disease treatment.
Collapse
Affiliation(s)
- Lei Sun
- George Washington University Cancer Center, USA; Department of Biochemistry & Molecular Medicine, George Washington University School of Medicine & Health Sciences, Washington, DC 20037, USA
| | | | - Ka Bian
- George Washington University Cancer Center, USA; Department of Biochemistry & Molecular Medicine, George Washington University School of Medicine & Health Sciences, Washington, DC 20037, USA
| | - Alexandra Achille
- Moffitt Cancer Center, University of South Florida, Tampa, FL 33612, USA
| | | | - Huadong Pei
- George Washington University Cancer Center, USA; Department of Biochemistry & Molecular Medicine, George Washington University School of Medicine & Health Sciences, Washington, DC 20037, USA
| | - Edward Seto
- George Washington University Cancer Center, USA; Department of Biochemistry & Molecular Medicine, George Washington University School of Medicine & Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|
22
|
Zhan Y, He Z, Liu X, Miao N, Lin F, Xu W, Mu S, Mu H, Yuan M, Cao X, Jin H, Liu Z, Li Y, Zhang B. Aspirin-induced attenuation of adipogenic differentiation of bone marrow mesenchymal stem cells is accompanied by the disturbed epigenetic modification. Int J Biochem Cell Biol 2018; 98:29-42. [DOI: 10.1016/j.biocel.2018.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/05/2018] [Accepted: 02/13/2018] [Indexed: 12/15/2022]
|
23
|
Basoli V, Santaniello S, Cruciani S, Ginesu GC, Cossu ML, Delitala AP, Serra PA, Ventura C, Maioli M. Melatonin and Vitamin D Interfere with the Adipogenic Fate of Adipose-Derived Stem Cells. Int J Mol Sci 2017; 18:ijms18050981. [PMID: 28475114 PMCID: PMC5454894 DOI: 10.3390/ijms18050981] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 04/28/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) represent one of the cellular populations resident in adipose tissue. They can be recruited under certain stimuli and committed to become preadipocytes, and then mature adipocytes. Controlling stem cell differentiation towards the adipogenic phenotype could have a great impact on future drug development aimed at counteracting fat depots. Stem cell commitment can be influenced by different molecules, such as melatonin, which we have previously shown to be an osteogenic inducer. Here, we aimed at evaluating the effects elicited by melatonin, even in the presence of vitamin D, on ADSC adipogenesis assessed in a specific medium. The transcription of specific adipogenesis orchestrating genes, such as aP2, peroxisome proliferator-activated receptor γ (PPAR-γ), and that of adipocyte-specific genes, including lipoprotein lipase (LPL) and acyl-CoA thioesterase 2 (ACOT2), was significantly inhibited in cells that had been treated in the presence of melatonin and vitamin D, alone or in combination. Protein content and lipid accumulation confirmed a reduction in adipogenesis in ADSCs that had been grown in adipogenic conditions, but in the presence of melatonin and/or vitamin D. Our findings indicate the role of melatonin and vitamin D in deciding stem cell fate, and disclose novel therapeutic approaches against fat depots.
Collapse
Affiliation(s)
- Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, A-1190 Vienna, Austria.
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, A-1190 Vienna, Austria.
| | - Giorgio Carlo Ginesu
- Clinical and Experimental Medicine Department, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
| | - Maria Laura Cossu
- Clinical and Experimental Medicine Department, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
| | | | - Pier Andrea Serra
- Clinical and Experimental Medicine Department, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
- Stem Wave Institute for Tissue Healing (SWITH), Gruppo VillaMaria and Ettore Sansavini Health Science Foundation, Via Provinciale per Cotignola 9, 48022 Lugo, Ravenna, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, Via Massarenti, 40138 Bologna, Italy.
- Stem Wave Institute for Tissue Healing (SWITH), Gruppo VillaMaria and Ettore Sansavini Health Science Foundation, Via Provinciale per Cotignola 9, 48022 Lugo, Ravenna, Italy.
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, 09042 Cagliari, Italy.
| |
Collapse
|
24
|
Yiew NKH, Chatterjee TK, Tang YL, Pellenberg R, Stansfield BK, Bagi Z, Fulton DJ, Stepp DW, Chen W, Patel V, Kamath VM, Litwin SE, Hui DY, Rudich SM, Kim HW, Weintraub NL. A novel role for the Wnt inhibitor APCDD1 in adipocyte differentiation: Implications for diet-induced obesity. J Biol Chem 2017; 292:6312-6324. [PMID: 28242765 PMCID: PMC5391760 DOI: 10.1074/jbc.m116.758078] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/15/2017] [Indexed: 01/03/2023] Open
Abstract
Impaired adipogenic differentiation during diet-induced obesity (DIO) promotes adipocyte hypertrophy and inflammation, thereby contributing to metabolic disease. Adenomatosis polyposis coli down-regulated 1 (APCDD1) has recently been identified as an inhibitor of Wnt signaling, a key regulator of adipogenic differentiation. Here we report a novel role for APCDD1 in adipogenic differentiation via repression of Wnt signaling and an epigenetic linkage between miR-130 and APCDD1 in DIO. APCDD1 expression was significantly up-regulated in mature adipocytes compared with undifferentiated preadipocytes in both human and mouse subcutaneous adipose tissues. siRNA-based silencing of APCDD1 in 3T3-L1 preadipocytes markedly increased the expression of Wnt signaling proteins (Wnt3a, Wnt5a, Wnt10b, LRP5, and β-catenin) and inhibited the expression of adipocyte differentiation markers (CCAAT/enhancer-binding protein α (C/EBPα) and peroxisome proliferator-activated receptor γ (PPARγ)) and lipid droplet accumulation, whereas adenovirus-mediated overexpression of APCDD1 enhanced adipogenic differentiation. Notably, DIO mice exhibited reduced APCDD1 expression and increased Wnt expression in both subcutaneous and visceral adipose tissues and impaired adipogenic differentiation in vitro Mechanistically, we found that miR-130, whose expression is up-regulated in adipose tissues of DIO mice, could directly target the 3'-untranslated region of the APCDD1 gene. Furthermore, transfection of an miR-130 inhibitor in preadipocytes enhanced, whereas an miR-130 mimic blunted, adipogenic differentiation, suggesting that miR-130 contributes to impaired adipogenic differentiation during DIO by repressing APCDD1 expression. Finally, human subcutaneous adipose tissues isolated from obese individuals exhibited reduced expression of APCDD1, C/EBPα, and PPARγ compared with those from non-obese subjects. Taken together, these novel findings suggest that APCDD1 positively regulates adipogenic differentiation and that its down-regulation by miR-130 during DIO may contribute to impaired adipogenic differentiation and obesity-related metabolic disease.
Collapse
Affiliation(s)
- Nicole K H Yiew
- From the Departments of Pharmacology and Toxicology
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Tapan K Chatterjee
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Medicine, Division of Cardiology
| | - Yao Liang Tang
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Medicine, Division of Cardiology
| | | | - Brian K Stansfield
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Pediatrics
| | - Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Medicine, Division of Cardiology
| | - David J Fulton
- From the Departments of Pharmacology and Toxicology
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - David W Stepp
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Physiology
| | | | | | | | - Sheldon E Litwin
- the Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - David Y Hui
- the Department of Pathology and Lab Medicine, University of Cincinnati, Cincinnati, Ohio 45219, and
| | | | - Ha Won Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912,
- Medicine, Division of Cardiology
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912,
- Medicine, Division of Cardiology
| |
Collapse
|
25
|
Plikus MV, Guerrero-Juarez CF, Ito M, Li YR, Dedhia PH, Zheng Y, Shao M, Gay DL, Ramos R, Hsi TC, Oh JW, Wang X, Ramirez A, Konopelski SE, Elzein A, Wang A, Supapannachart RJ, Lee HL, Lim CH, Nace A, Guo A, Treffeisen E, Andl T, Ramirez RN, Murad R, Offermanns S, Metzger D, Chambon P, Widgerow AD, Tuan TL, Mortazavi A, Gupta RK, Hamilton BA, Millar SE, Seale P, Pear WS, Lazar MA, Cotsarelis G. Regeneration of fat cells from myofibroblasts during wound healing. Science 2017; 355:748-752. [PMID: 28059714 DOI: 10.1126/science.aai8792] [Citation(s) in RCA: 387] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/19/2016] [Indexed: 12/14/2022]
Abstract
Although regeneration through the reprogramming of one cell lineage to another occurs in fish and amphibians, it has not been observed in mammals. We discovered in the mouse that during wound healing, adipocytes regenerate from myofibroblasts, a cell type thought to be differentiated and nonadipogenic. Myofibroblast reprogramming required neogenic hair follicles, which triggered bone morphogenetic protein (BMP) signaling and then activation of adipocyte transcription factors expressed during development. Overexpression of the BMP antagonist Noggin in hair follicles or deletion of the BMP receptor in myofibroblasts prevented adipocyte formation. Adipocytes formed from human keloid fibroblasts either when treated with BMP or when placed with human hair follicles in vitro. Thus, we identify the myofibroblast as a plastic cell type that may be manipulated to treat scars in humans.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA. .,Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Yun Rose Li
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Priya H Dedhia
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Zheng
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Denise L Gay
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,INSERM U967, Commissariat à L'énergie Atomique et aux Énergies Alternatives, Institut de Radiobiologie Cellulaire et Moléculaire 92265 Fontenay-aux-Roses Cedex, France
| | - Raul Ramos
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Won Oh
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.,Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Amanda Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Sara E Konopelski
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Arijh Elzein
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Anne Wang
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rarinthip June Supapannachart
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hye-Lim Lee
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Arben Nace
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amy Guo
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elsa Treffeisen
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 328116, USA
| | - Ricardo N Ramirez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rabi Murad
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Daniel Metzger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Université de Strasbourg, Illkirch 67404, France
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, INSERM U964, Institut d'Etudes Avancées de l'Université de Strasbourg, Collège de France, Illkirch 67404, France
| | - Alan D Widgerow
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, Irvine, CA 92868, USA
| | - Tai-Lan Tuan
- The Saban Research Institute of Children's Hospital Los Angeles and Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bruce A Hamilton
- Departments of Medicine and Cellular and Molecular Medicine, Moores Cancer Center and Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sarah E Millar
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Seale
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Kligman Laboratories, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Abstract
BACKGROUND: Skeletal muscle atrophy during aging, a process known as sarcopenia, is associated with muscle weakness, frailty, and the loss of independence in older adults. The mechanisms contributing to sarcopenia are not totally understood, but muscle fiber loss due to apoptosis, reduced stimulation of anabolic pathways, activation of catabolic pathways, denervation, and altered metabolism have been observed in muscle from old rodents and humans. OBJECTIVE: Recently, histone deacetylases (HDACs) have been implicated in muscle atrophy and dysfunction due to denervation, muscular dystrophy, and disuse, and HDACs play key roles in regulating metabolism in skeletal muscle. In this review, we will discuss the role of HDACs in muscle atrophy and the potential of HDAC inhibitors for the treatment of sarcopenia. CONCLUSIONS: Several HDAC isoforms are potential targets for intervention in sarcopenia. Inhibition of HDAC1 prevents muscle atrophy due to nutrient deprivation. HDAC3 regulates metabolism in skeletal muscle and may inhibit oxidative metabolism during aging. HDAC4 and HDAC5 have been implicated in muscle atrophy due to denervation, a process implicated in sarcopenia. HDAC inhibitors are already in use in the clinic, and there is promise in targeting HDACs for the treatment of sarcopenia.
Collapse
Affiliation(s)
- Michael E Walsh
- Energy Metabolism Laboratory, Swiss Federal Institute of Technology (ETH) Zurich , Zurich, Switzerland
| | | |
Collapse
|
27
|
Cheung OKW, Cheng ASL. Gender Differences in Adipocyte Metabolism and Liver Cancer Progression. Front Genet 2016; 7:168. [PMID: 27703473 PMCID: PMC5029146 DOI: 10.3389/fgene.2016.00168] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022] Open
Abstract
Liver cancer is the third most common cancer type and the second leading cause of deaths in men. Large population studies have demonstrated remarkable gender disparities in the incidence and the cumulative risk of liver cancer. A number of emerging risk factors regarding metabolic alterations associated with obesity, diabetes and dyslipidemia have been ascribed to the progression of non-alcoholic fatty liver diseases (NAFLD) and ultimately liver cancer. The deregulation of fat metabolism derived from excessive insulin, glucose, and lipid promotes cancer-causing inflammatory signaling and oxidative stress, which eventually triggers the uncontrolled hepatocellular proliferation. This review presents the current standing on the gender differences in body fat compositions and their mechanistic linkage with the development of NAFLD-related liver cancer, with an emphasis on genetic, epigenetic and microRNA control. The potential roles of sex hormones in instructing adipocyte metabolic programs may help unravel the mechanisms underlying gender dimorphism in liver cancer and identify the metabolic targets for disease management.
Collapse
Affiliation(s)
- Otto K-W Cheung
- School of Biomedical Sciences, The Chinese University of Hong Kong Hong Kong, China
| | - Alfred S-L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong Hong Kong, China; State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong Hong Kong, China
| |
Collapse
|
28
|
Liu F, Zong M, Wen X, Li X, Wang J, Wang Y, Jiang W, Li X, Guo Z, Qi H. Silencing of Histone Deacetylase 9 Expression in Podocytes Attenuates Kidney Injury in Diabetic Nephropathy. Sci Rep 2016; 6:33676. [PMID: 27633396 PMCID: PMC5025656 DOI: 10.1038/srep33676] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 08/31/2016] [Indexed: 12/21/2022] Open
Abstract
Podocyte dysfunction is important in the onset and development of diabetic nephropathy (DN). Histone deacetylases (HDACs) have been recently proved to play critical roles in the pathogenesis of DN. As one subtype of the class IIa HDACs, HDAC9 is capable to repress/de-repress their target genes in tumor, inflammation, atherosclerosis and metabolic diseases. In the present study, we investigate whether HDAC9 is involved in the pathophysiologic process of DN, especially the podocyte injury. Firstly, we explored the expression patterns and localization of HDAC9 and found that HDAC9 expression was significantly up-regulated in high glucose (HG)-treated mouse podocytes, as well as kidney tissues from diabetic db/db mice and patients with DN. Secondly, knockdown of HDAC9 in mouse podocytes significantly suppressed HG-induced reactive oxygen species (ROS) generation, cell apoptosis and inflammation through JAK2/STAT3 pathway and reduced the podocytes injury by decreasing the expression levels of Nephrin and Podocin. Moreover, in diabetic db/db mice, silencing of HDAC9 attenuated the glomerulosclerosis, inflammatory cytokine release, podocyte apoptosis and renal injury. Collectively, these data indicate that HDAC9 may be involved in the process of DN, especially podocyte injury. Our study suggest that inhibition of HDAC9 may have a therapeutic potential in DN treatment.
Collapse
Affiliation(s)
- Feng Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ming Zong
- Department of Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaofei Wen
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuezhu Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Jiang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaojun Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongliang Guo
- Department of Respiratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hualin Qi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Sharma S, Taliyan R. Epigenetic modifications by inhibiting histone deacetylases reverse memory impairment in insulin resistance induced cognitive deficit in mice. Neuropharmacology 2016; 105:285-297. [PMID: 26805421 DOI: 10.1016/j.neuropharm.2016.01.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/12/2016] [Accepted: 01/20/2016] [Indexed: 01/04/2023]
Abstract
Insulin resistance has been reported as a strong risk factor for Alzheimer's disease. However the molecular mechanisms of association between these still remain elusive. Various studies have highlighted the involvement of histone deacetylases (HDACs) in insulin resistance and cognitive deficits. Thus, the present study was designed to investigate the possible neuroprotective role of HDAC inhibitor, suberoylanilide hydroxamic acid (SAHA) in insulin resistance induced cognitive impairment in mice. Mice were subjected to either normal pellet diet (NPD) or high fat diet (HFD) for 8 weeks. HFD fed mice were treated with SAHA at 25 and 50 mg/kg i.p. once daily for 2 weeks. Serum insulin, glucose, triglycerides, total cholesterol and HDL-cholesterol levels were measured. A battery of behavioral parameters was performed to assess cognitive functions. Level of tumour necrosis factor (TNF-α) was measured in hippocampus to assess neuroinflammation. To further explore the molecular mechanisms we measured the histone H3 acetylation and brain derived neurotrophic factor (BDNF) level. HFD fed mice exhibit characteristic features of insulin resistance. These mice also showed a severe deficit in learning and memory along with reduced histone H3 acetylation and BDNF levels. In contrast, the mice treated with SAHA showed significant and dose dependent improvement in insulin resistant condition. These mice also showed improved learning and memory performance. SAHA treatment ameliorates the HFD induced reduction in histone H3 acetylation and BDNF levels. Based upon these results, it could be suggested that HDAC inhibitors exert neuroprotective effects by increasing H3 acetylation and subsequently BDNF level.
Collapse
Affiliation(s)
- Sorabh Sharma
- Neuropharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India
| | - Rajeev Taliyan
- Neuropharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India.
| |
Collapse
|