1
|
Wang W, Pang Z, Zhang S, Yang P, Pan Y, Qiao L, Yang K, Liu J, Wang R, Liu W. Multi-omics integrated analysis reveals the molecular mechanism of tail fat deposition differences in sheep with different tail types. BMC Genomics 2025; 26:465. [PMID: 40346476 PMCID: PMC12065285 DOI: 10.1186/s12864-025-11658-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND The accumulation of tail fat in sheep is a manifestation of adaptive evolution to the environment. Sheep with different tail types show significant differences in physiological functions and tail fat deposition. Although these differences reflect the developmental mechanism of tail fat under different gene regulation, the situation of sheep tail fat tissue at the single cell level has not been explored, and its molecular mechanism still needs to be further elucidated. RESULTS Here, we characterized the genomic features of sheep with different tail types, detected the transcriptomic differences in tail adipose tissue between fat-tailed and thin-tailed sheep, established a single-cell atlas of sheep tail adipose tissue, and screened potential molecular markers (SESN1, RPRD1A and RASGEF1B) that regulate differences in sheep tail fat deposition through multi-omics integrated analysis. We found that the differential mechanism of sheep tail fat deposition not only involves adipocyte differentiation and proliferation, but is also closely related to cell-specific communication networks (When adipocytes act as signal outputters, LAMININ and other signal pathways are strongly expressed in guangling large tailed sheep and hu sheep), including interactions with immune cells and tissue remodeling to drive the typing of tail fat. In addition, we revealed the differentiation trajectory of sheep tail adipocytes through pseudo-time analysis and constructed the cell communication network of sheep tail adipose tissue. CONCLUSIONS Our results provide insights into the molecular mechanisms of tail fat deposition in sheep with different tail types, and provide a deeper explanation for the development and functional regulation of adipocytes.
Collapse
Affiliation(s)
- Wannian Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Zhixv Pang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Siying Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Pengkun Yang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Yangyang Pan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Liying Qiao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Kaijie Yang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Jianhua Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
- Key Laboratory of Farm Animal Genetic Resources Exploration and Precision Breeding of Shanxi Province, Taigu, 030801, China
| | - Ruizhen Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Wenzhong Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
- Key Laboratory of Farm Animal Genetic Resources Exploration and Precision Breeding of Shanxi Province, Taigu, 030801, China.
| |
Collapse
|
2
|
Minaya DM, Hoss A, Bhagat A, Guo TL, Czaja K. Sex-Specific Effect of a High-Energy Diet on Body Composition, Gut Microbiota, and Inflammatory Markers in Rats. Nutrients 2025; 17:1147. [PMID: 40218905 PMCID: PMC11990636 DOI: 10.3390/nu17071147] [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: 02/05/2025] [Revised: 03/16/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: A high-energy-density (HED) diet promotes body weight gain, fat accumulation, and gut dysbiosis, contributing to obesity. The aim of this study was to characterize the initial response to HED diet consumption, as well as identify any sex differences in body composition, systemic inflammation, gut microbiome, and fecal fat excretion in rats. Methods: Male and female Sprague-Dawley rats were fed a low-energy-density (LED) diet for 10 days and were then switched to an HED diet for four weeks. Food intake, body weight, and body composition were measured routinely. Serum samples were collected to measure inflammatory cytokines/chemokines. Fecal samples were collected for microbiome analysis and lipid content. Results: After the HED diet, all rats gained body weight and fat mass, with males exhibiting increased susceptibility to weight gain. Males displayed either a diet-induced obesity phenotype (DIO-P) or a diet-resistant (DR) phenotype, as characterized by their differential body weight gain. Males showed elevated TGF-β levels, while females exhibited increases in Interferon gamma-inducible protein 10 (IP-10), regulated on activation, normal T cell expressed and secreted (RANTES) protein, and basic fibroblast growth factor (FGFb). Changes in gut microbiota composition revealed a reduction in beneficial species, like Bacteroides uniformis and Parabacteroides distasonis, and an increase in species such as Akkermansia muciniphila. Sex differences in fat metabolism were shown in the greater fecal fat excretion observed in males. Conclusions: Our study demonstrates that short-term consumption of a high-energy diet elicits notable sex-specific differences in body weight, body composition, inflammatory markers, gut microbiota, and fat excretion in Sprague-Dawley rats. While we recognize that this study has a small sample size and a short-term intervention, our findings highlight the critical role of sex as a biological variable in diet-induced obesity research.
Collapse
Affiliation(s)
- Dulce M. Minaya
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA 30605, USA;
| | - Adam Hoss
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (A.H.); (T.L.G.)
| | - Ayushi Bhagat
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| | - Tai L. Guo
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (A.H.); (T.L.G.)
| | - Krzysztof Czaja
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (A.H.); (T.L.G.)
| |
Collapse
|
3
|
Listyoko AS, Okazaki R, Harada T, Inui G, Yamasaki A. Impact of obesity on airway remodeling in asthma: pathophysiological insights and clinical implications. FRONTIERS IN ALLERGY 2024; 5:1365801. [PMID: 38562155 PMCID: PMC10982419 DOI: 10.3389/falgy.2024.1365801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of obesity among asthma patients has surged in recent years, posing a significant risk factor for uncontrolled asthma. Beyond its impact on asthma severity and patients' quality of life, obesity is associated with reduced lung function, increased asthma exacerbations, hospitalizations, heightened airway hyperresponsiveness, and elevated asthma-related mortality. Obesity may lead to metabolic dysfunction and immune dysregulation, fostering chronic inflammation characterized by increased pro-inflammatory mediators and adipocytokines, elevated reactive oxygen species, and reduced antioxidant activity. This chronic inflammation holds the potential to induce airway remodeling in individuals with asthma and obesity. Airway remodeling encompasses structural and pathological changes, involving alterations in the airway's epithelial and subepithelial layers, hyperplasia and hypertrophy of airway smooth muscle, and changes in airway vascularity. In individuals with asthma and obesity, airway remodeling may underlie heightened airway hyperresponsiveness and increased asthma severity, ultimately contributing to the development of persistent airflow limitation, declining lung function, and a potential increase in asthma-related mortality. Despite efforts to address the impact of obesity on asthma outcomes, the intricate mechanisms linking obesity to asthma pathophysiology, particularly concerning airway remodeling, remain incompletely understood. This comprehensive review discusses current research investigating the influence of obesity on airway remodeling, to enhance our understanding of obesity's role in the context of asthma airway remodeling.
Collapse
Affiliation(s)
- Aditya Sri Listyoko
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
- Pulmonology and Respiratory Medicine Department, Faculty of Medicine, Brawijaya University-Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Genki Inui
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
4
|
Nunn E, Jaiswal N, Gavin M, Uehara K, Stefkovich M, Drareni K, Calhoun R, Lee M, Holman CD, Baur JA, Seale P, Titchenell PM. Antibody blockade of activin type II receptors preserves skeletal muscle mass and enhances fat loss during GLP-1 receptor agonism. Mol Metab 2024; 80:101880. [PMID: 38218536 PMCID: PMC10832506 DOI: 10.1016/j.molmet.2024.101880] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
OBJECTIVE Glucagon-like peptide 1 (GLP-1) receptor agonists reduce food intake, producing remarkable weight loss in overweight and obese individuals. While much of this weight loss is fat mass, there is also a loss of lean mass, similar to other approaches that induce calorie deficit. Targeting signaling pathways that regulate skeletal muscle hypertrophy is a promising avenue to preserve lean mass and modulate body composition. Myostatin and Activin A are TGFβ-like ligands that signal via the activin type II receptors (ActRII) to antagonize muscle growth. Pre-clinical and clinical studies demonstrate that ActRII blockade induces skeletal muscle hypertrophy and reduces fat mass. In this manuscript, we test the hypothesis that combined ActRII blockade and GLP-1 receptor agonism will preserve muscle mass, leading to improvements in skeletomuscular and metabolic function and enhanced fat loss. METHODS In this study, we explore the therapeutic potential of bimagrumab, a monoclonal antibody against ActRII, to modify body composition alone and during weight loss induced by GLP-1 receptor agonist semaglutide in diet-induced obese mice. Mechanistically, we define the specific role of the anabolic kinase Akt in mediating the hypertrophic muscle effects of ActRII inhibition in vivo. RESULTS Treatment of obese mice with bimagrumab induced a ∼10 % increase in lean mass while simultaneously decreasing fat mass. Daily treatment of obese mice with semaglutide potently decreased body weight; this included a significant decrease in both muscle and fat mass. Combination treatment with bimagrumab and semaglutide led to superior fat mass loss while simultaneously preserving lean mass despite reduced food intake. Treatment with both drugs was associated with improved metabolic outcomes, and increased lean mass was associated with improved exercise performance. Deletion of both Akt isoforms in skeletal muscle modestly reduced, but did not prevent, muscle hypertrophy driven by ActRII inhibition. CONCLUSIONS Collectively, these data demonstrate that blockade of ActRII signaling improves body composition and metabolic parameters during calorie deficit driven by GLP-1 receptor agonism and demonstrate the existence of Akt-independent pathways supporting muscle hypertrophy in the absence of ActRII signaling.
Collapse
Affiliation(s)
- Elizabeth Nunn
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Natasha Jaiswal
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Gavin
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kahealani Uehara
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Megan Stefkovich
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Karima Drareni
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Calhoun
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michelle Lee
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Corey D Holman
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Kar A, Alvarez M, Garske KM, Huang H, Lee SHT, Deal M, Das SS, Koka A, Jamal Z, Mohlke KL, Laakso M, Heinonen S, Pietiläinen KH, Pajukanta P. Age-dependent genes in adipose stem and precursor cells affect regulation of fat cell differentiation and link aging to obesity via cellular and genetic interactions. Genome Med 2024; 16:19. [PMID: 38297378 PMCID: PMC10829214 DOI: 10.1186/s13073-024-01291-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/19/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Age and obesity are dominant risk factors for several common cardiometabolic disorders, and both are known to impair adipose tissue function. However, the underlying cellular and genetic factors linking aging and obesity on adipose tissue function have remained elusive. Adipose stem and precursor cells (ASPCs) are an understudied, yet crucial adipose cell type due to their deterministic adipocyte differentiation potential, which impacts the capacity to store fat in a metabolically healthy manner. METHODS We integrated subcutaneous adipose tissue (SAT) bulk (n=435) and large single-nucleus RNA sequencing (n=105) data with the UK Biobank (UKB) (n=391,701) data to study age-obesity interactions originating from ASPCs by performing cell-type decomposition, differential expression testing, cell-cell communication analyses, and construction of polygenic risk scores for body mass index (BMI). RESULTS We found that the SAT ASPC proportions significantly decrease with age in an obesity-dependent way consistently in two independent cohorts, both showing that the age dependency of ASPC proportions is abolished by obesity. We further identified 76 genes (72 SAT ASPC marker genes and 4 transcription factors regulating ASPC marker genes) that are differentially expressed by age in SAT and functionally enriched for developmental processes and adipocyte differentiation (i.e., adipogenesis). The 76 age-perturbed ASPC genes include multiple negative regulators of adipogenesis, such as RORA, SMAD3, TWIST2, and ZNF521, form tight clusters of longitudinally co-expressed genes during human adipogenesis, and show age-based differences in cellular interactions between ASPCs and adipose cell types. Finally, our genetic data demonstrate that cis-regional variants of these genes interact with age as predictors of BMI in an obesity-dependent way in the large UKB, while no such gene-age interaction on BMI is observed with non-age-dependent ASPC marker genes, thus independently confirming our cellular ASPC results at the biobank level. CONCLUSIONS Overall, we discover that obesity prematurely induces a decrease in ASPC proportions and identify 76 developmentally important ASPC genes that implicate altered negative regulation of fat cell differentiation as a mechanism for aging and directly link aging to obesity via significant cellular and genetic interactions.
Collapse
Affiliation(s)
- Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Huiling Huang
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA
| | - Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Milena Deal
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Sankha Subhra Das
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Zoeb Jamal
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA.
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA.
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, USA.
| |
Collapse
|
6
|
Wang F, Xu SJ, Ye F, Zhang B, Sun XB. Integration of Transcriptomics and Lipidomics Profiling to Reveal the Therapeutic Mechanism Underlying Ramulus mori (Sangzhi) Alkaloids for the Treatment of Liver Lipid Metabolic Disturbance in High-Fat-Diet/Streptozotocin-Induced Diabetic Mice. Nutrients 2023; 15:3914. [PMID: 37764698 PMCID: PMC10536214 DOI: 10.3390/nu15183914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder, with a global prevalence of 25%. Currently, there remains no approved therapy. Ramulus mori (Sangzhi) alkaloids (SZ-As), a novel natural medicine, have achieved comprehensive benefits in the treatment of type 2 diabetes; however, few studies have focused on its role in ameliorating hepatic lipid metabolic disturbance. Herein, the therapeutic effect and mechanism of SZ-As on a high-fat diet (HFD) combined with streptozotocin (STZ)-induced NAFLD mice were investigated via incorporating transcriptomics and lipidomics. SZ-As reduced body weight and hepatic lipid levels, restored pathological alternation and converted the blood biochemistry perturbations. SZ-A treatment also remarkedly inhibited lipogenesis and enhanced lipolysis, fatty acid oxidation and thermogenesis. Transcriptomics analysis confirmed that SZ-As mainly altered fatty acid oxidative metabolism and the TNF signaling pathway. SZ-As were further demonstrated to downregulate inflammatory factors and effectively ameliorate hepatic inflammation. Lipidomics analysis also suggested that SZ-As affected differential lipids including triglyceride (TG) and phosphatidylcholine (PC) expression, and the main metabolic pathways included glycerophospholipid, sphingomyelins and choline metabolism. Collectively, combined with transcriptomics and metabolomics data, it is suggested that SZ-As exert their therapeutic effect on NAFLD possibly through regulating lipid metabolism pathways (glycerophospholipid metabolism and choline metabolism) and increasing levels of PC and lysophosphatidylcholine (LPC) metabolites. This study provides the basis for more widespread clinical applications of SZ-As.
Collapse
Affiliation(s)
- Fan Wang
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; (F.W.); (S.-J.X.); (F.Y.)
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Sai-Jun Xu
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; (F.W.); (S.-J.X.); (F.Y.)
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Fan Ye
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; (F.W.); (S.-J.X.); (F.Y.)
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; (F.W.); (S.-J.X.); (F.Y.)
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| | - Xiao-Bo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100193, China; (F.W.); (S.-J.X.); (F.Y.)
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
| |
Collapse
|
7
|
Ren L, Liu X, Huang X, Zhang H, Fei W, Yu X, Hu Z, Zhen Y, Chen S. Oxymatrine relieves high-fructose/fat-induced obesity via reprogramming the activity of lipid metabolism-related enhancer. Front Endocrinol (Lausanne) 2023; 14:1145575. [PMID: 37600712 PMCID: PMC10437059 DOI: 10.3389/fendo.2023.1145575] [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: 01/16/2023] [Accepted: 04/11/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Emerging evidence demonstrates that the high-fructose and high-fat diet (HFHF) induced obesity and fatty liver disease has become one of the most common metabolic disorders worldwide. Therefore, innovative investigations on compounds targeting obesity and fatty liver diseases are urgently needed. Methods The high-throughput natural compounds screen was performed to screen the important compounds. A rat HFHF model was constructed, the regulatory function of Oxymatrine in HFHF-induced obesity was further explored. Results We identified Oxymatrine, a natural compound extracted from Sophora flavescens, showed a potential compacity in high-fat diet-induced fatty liver disease. We found that oxymatrine significantly inhibited HFHF-induced obesity using a rat HFHF model. Additionally, we found that oxymatrine altered the enhancer landscape of subcutaneous adipose tissues by ChIP-seq analysis using antibodies against the H3K27ac histone modification. Motif enrichment analysis showed the Smad motif was significantly enriched in enhancers altered post-oxymatrine treatment. Further chromatin immunoprecipitation-quantitative PCR (ChIP-qPCR) analysis and luciferase reporter assays showed oxymatrine alters the binding of Smad3 on the enhancer regions of B-cell lymphoma 2 (Bcl2) and the enhancer activity of Bcl2. Discussion Together, our study highlighted oxymatrine could suppress high-fructose and high-fat diet-induced obesity by inhibiting the suppressor of mothers against decapentaplegic 3 (Smad3) binding on obesity-related enhancers.
Collapse
Affiliation(s)
- Luping Ren
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xuehua Liu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
- Graduate School of Hebei North University, Zhangjiakou, Hebei, China
| | - Xitong Huang
- Department of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - He Zhang
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Wenjie Fei
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xian Yu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Zhijuan Hu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yunfeng Zhen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
8
|
Yang M, Zhang R, Liu X, Shi G, Liu H, Wang L, Hou X, Shi L, Wang L, Zhang L. Integrating genome-wide association study with RNA-seq revealed DBI as a good candidate gene for intramuscular fat content in Beijing black pigs. Anim Genet 2023; 54:24-34. [PMID: 36305366 DOI: 10.1111/age.13270] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 10/04/2022] [Accepted: 10/08/2022] [Indexed: 01/07/2023]
Abstract
Increasing intramuscular fat (IMF) content can enhance the sensory quality of meat, including tenderness, juiciness, flavor, and color. Genome-wide association study and RNA-sequencing (RNA-seq) analysis were used to identify candidate IMF genes in Beijing Black pigs, a popular species among consumers in northern China. Two and three single nucleotide polymorphisms were significantly associated with IMF in SSC13 and SSC15 respectively. Solute carrier family 4 member 7 (SLC4A7) on SSC13 and insulin induced gene 2 (INSIG2), coiled-coil domain containing 93 (CCDC93), and diazepam binding inhibitor acyl-CoA binding protein (DBI) on SSC15 are good candidate genes in this population. Furthermore, RNA-seq analysis was performed between high and low IMF groups, and 534 differentially expressed genes were identified. In addition, based on differentially expressed genes, Kyoto Encyclopedia of Genes and Genomes analysis revealed that peroxisome proliferator-activated receptors and FoxO signaling pathway pathways might contribute to IMF. Moreover, the DBI gene was identified as a candidate for IMF both by genome-wide association study and RNA-seq analysis, suggesting that it might be a crucial candidate gene for influencing IMF in Beijing Black pigs.
Collapse
Affiliation(s)
- Man Yang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Run Zhang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiance Liu
- Beijing Heiliu Animal Husbandry Technology Co, Ltd, Beijing, China
| | - Guohua Shi
- Beijing Heiliu Animal Husbandry Technology Co, Ltd, Beijing, China
| | - Hai Liu
- Beijing Heiliu Animal Husbandry Technology Co, Ltd, Beijing, China
| | - Ligang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xinhua Hou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lijun Shi
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lixian Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Longchao Zhang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
9
|
Di Cicco M, Ghezzi M, Kantar A, Song WJ, Bush A, Peroni D, D'Auria E. Pediatric obesity and severe asthma: Targeting pathways driving inflammation. Pharmacol Res 2023; 188:106658. [PMID: 36642111 DOI: 10.1016/j.phrs.2023.106658] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Asthma affects more than 300 million people of all ages worldwide, including about 10-15% of school-aged children, and its prevalence is increasing. Severe asthma (SA) is a particular and rare phenotype requiring treatment with high-dose inhaled corticosteroids plus a second controller and/or systemic glucocorticoid courses to achieve symptom control or remaining "uncontrolled" despite this therapy. In SA, other diagnoses have been excluded, and potential exacerbating factors have been addressed. Notably, obese asthmatics are at higher risk of developing SA. Obesity is both a major risk factor and a disease modifier of asthma in children and adults: two main "obese asthma" phenotypes have been described in childhood with high or low levels of Type 2 inflammation biomarkers, respectively, the former characterized by early onset and eosinophilic inflammation and the latter by neutrophilic inflammation and late-onset. Nevertheless, the interplay between obesity and asthma is far more complex and includes obese tissue-driven inflammatory pathways, mechanical factors, comorbidities, and poor response to corticosteroids. This review outlines the most recent findings on SA in obese children, particularly focusing on inflammatory pathways, which are becoming of pivotal importance in order to identify selective targets for specific treatments, such as biological agents.
Collapse
Affiliation(s)
- Maria Di Cicco
- Pediatric Clinic, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michele Ghezzi
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Ahmad Kantar
- Pediatric Asthma and Cough Centre, Gruppo Ospedaliero San Donato, Bergamo, Italy and Università Vita Salute San Raffaele, Milan, Italy
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Andrew Bush
- Dept of Paediatric Respiratory Medicine, Royal Brompton Hospital and National Heart and Lung Institute, School of Medicine, Imperial College London, London, UK
| | - Diego Peroni
- Pediatric Clinic, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Enza D'Auria
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy.
| |
Collapse
|
10
|
Zou L, Feng Q, Xia W, Zhu C. Bioinformatics analysis of the common targets of miR-223-3p, miR-122-5p, and miR-93-5p in polycystic ovarian syndrome. Front Genet 2023; 14:1097706. [PMID: 36873932 PMCID: PMC9977968 DOI: 10.3389/fgene.2023.1097706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is one of the most common gynecological endocrine disorders. MicroRNAs (miRNAs) play extensive roles in the pathogenesis of PCOS and can serve as potential diagnostic markers. However, most studies focused on the regulatory mechanisms of individual miRNAs, and the combined regulatory effects of multiple miRNAs remain unclear. The aim of this study was to identify the common targets of miR-223-3p, miR-122-5p, and miR-93-5p; and assess the transcript levels of some of these targets in PCOS rat ovaries. Transcriptome profiles of granulosa cells from PCOS patients were obtained from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs). A total of 1,144 DEGs were screened, 204 of which were upregulated and 940 were downregulated. According to the miRWalk algorithm, 4,284 genes were targeted by all three miRNAs at the same time, and intersection with DEGs was used to obtain candidate target genes. A total of 265 candidate target genes were screened, and the detected target genes were subjected to Gene ontology (GO) and KEGG pathway enrichment, followed by PPI network analysis. Then, qRT-PCR was used to determine the levels of 12 genes in PCOS rat ovaries. The expressions of 10 of these genes were found to be consistent with our bioinformatics results. In conclusion, JMJD1C, PLCG2, SMAD3, FOSL2, TGFB1, TRIB1, GAS7, TRIM25, NFYA, and CALCRL may participate in the development of PCOS. Our findings contribute to the identification of biomarkers that may promote the effective prevention and treatment of PCOS in the future.
Collapse
Affiliation(s)
- Liping Zou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiwen Feng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xia
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changhong Zhu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Kaushik S, Juste YR, Lindenau K, Dong S, Macho-González A, Santiago-Fernández O, McCabe M, Singh R, Gavathiotis E, Cuervo AM. Chaperone-mediated autophagy regulates adipocyte differentiation. SCIENCE ADVANCES 2022; 8:eabq2733. [PMID: 36383673 PMCID: PMC9668314 DOI: 10.1126/sciadv.abq2733] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
Adipogenesis is a tightly orchestrated multistep process wherein preadipocytes differentiate into adipocytes. The most studied aspect of adipogenesis is its transcriptional regulation through timely expression and silencing of a vast number of genes. However, whether turnover of key regulatory proteins per se controls adipogenesis remains largely understudied. Chaperone-mediated autophagy (CMA) is a selective form of lysosomal protein degradation that, in response to diverse cues, remodels the proteome for regulatory purposes. We report here the activation of CMA during adipocyte differentiation and show that CMA regulates adipogenesis at different steps through timely degradation of key regulatory signaling proteins and transcription factors that dictate proliferation, energetic adaptation, and signaling changes required for adipogenesis.
Collapse
Affiliation(s)
- Susmita Kaushik
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yves R. Juste
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kristen Lindenau
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shuxian Dong
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Adrián Macho-González
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Olaya Santiago-Fernández
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mericka McCabe
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rajat Singh
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evripidis Gavathiotis
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
12
|
Das R, Giri J, K Paul P, Froelich N, Chinnadurai R, McCoy S, Bushman W, Galipeau J. A STAT5-Smad3 dyad regulates adipogenic plasticity of visceral adipose mesenchymal stromal cells during chronic inflammation. NPJ Regen Med 2022; 7:41. [PMID: 36045134 PMCID: PMC9433418 DOI: 10.1038/s41536-022-00244-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 08/10/2022] [Indexed: 11/27/2022] Open
Abstract
Adipogenic differentiation of visceral adipose tissue-resident multipotent mesenchymal stromal cells (VA-MSC) into adipocytes is metabolically protective. Under chronic inflammatory stress, this neoadipogenesis process is suppressed by various pro-inflammatory cytokines and growth factors. However, the underlying mechanism(s) regulating VA-MSC plasticity remains largely unexplored. Using an adipogenic differentiation screen, we identified IFNγ and TGFβ as key inhibitors of primary human VA-MSC differentiation. Further studies using human and mouse VA-MSCs and a chronic high-fat diet-fed murine model revealed that IFNγ/JAK2-activated STAT5 transcription factor is a central regulator of VA-MSC differentiation under chronic inflammatory conditions. Furthermore, our results indicate that under such conditions, IFNγ-activated STAT5 and TGFβ-activated Smad3 physically interact via Smad4. This STAT5-Smad4-Smad3 complex plays a crucial role in preventing the early adipogenic commitment of VA-MSCs by suppressing key pro-adipogenic transcription factors, including CEBPδ, CEBPα, and PPARγ. Genetic or pharmacological disruption of IFNγ-TGFβ synergy by inhibiting either STAT5 or Smad3 rescued adipogenesis under chronic inflammatory stress. Overall, our study delineates a central mechanism of MSC plasticity regulation by the convergence of multiple inflammatory signaling pathways.
Collapse
Affiliation(s)
- Rahul Das
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jayeeta Giri
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Pradyut K Paul
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Nicole Froelich
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Raghavan Chinnadurai
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
- School of Medicine, Mercer University, Savannah, GA, 31404, USA
| | - Sara McCoy
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Wade Bushman
- Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jacques Galipeau
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
13
|
Crouse WL, Das SK, Le T, Keele G, Holl K, Seshie O, Craddock A, Sharma NK, Comeau ME, Langefeld C, Hawkins GA, Mott R, Valdar W, Solberg Woods LC. Transcriptome-wide analyses of adipose tissue in outbred rats reveal genetic regulatory mechanisms relevant for human obesity. Physiol Genomics 2022; 54:206-219. [PMID: 35467982 DOI: 10.1152/physiolgenomics.00172.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transcriptomic analysis in metabolically active tissues allows a systems genetics approach to identify causal genes and networks involved in metabolic disease. Outbred heterogeneous stock (HS) rats are used for genetic mapping of complex traits, but to-date, a systems genetics analysis of metabolic tissues has not been done. We investigated whether adiposity-associated genes and gene co-expression networks in outbred heterogeneous stock (HS) rats overlap those found in humans. We analyzed RNAseq data from adipose tissue of 415 male HS rats, correlated these transcripts with body weight (BW) and compared transcriptome signatures to two human cohorts: the "African American Genetics of Metabolism and Expression" and "Metabolic Syndrome in Men". We used weighted gene co-expression network analysis to identify adiposity-associated gene networks and mediation analysis to identify genes under genetic control whose expression drives adiposity. We identified 554 orthologous "consensus genes" whose expression correlates with BW in the rat and with body mass index (BMI) in both human cohorts. Consensus genes fell within eight co-expressed networks and were enriched for genes involved in immune system function, cell growth, extracellular matrix organization and lipid metabolic processes. We identified 19 consensus genes for which genetic variation may influence BW via their expression, including those involved in lipolysis (e.g., Hcar1), inflammation (e.g., Rgs1), adipogenesis (e.g., Tmem120b) or no previously known role in obesity (e.g., St14, Msa4a6). Strong concordance between HS rat and human BW/BMI associated transcripts demonstrates translational utility of the rat model, while identification of novel genes expands our knowledge of the genetics underlying obesity.
Collapse
Affiliation(s)
- Wesley L Crouse
- University of North Carolina at Chapel Hill, Department of Genetics, Chapel Hill, NC, United States
| | - Swapan Kumar Das
- Wake Forest University School of Medicine, Department of Internal Medicine, Winston Salem, NC, United States
| | - Thu Le
- University College London, Department of Genetics, Evolution and Environment, Division of Biosciences, London, United Kingdom
| | - Gregory Keele
- Jackson Laboratories, Roux Center for Genomics and Computational Biology, Bar Harbor, ME, United States
| | - Katie Holl
- Medical College of Wisconsin, Department of Pediatrics, Milwaukee, WI, United States
| | - Osborne Seshie
- Wake Forest University School of Medicine, Department of Internal Medicine, Winston Salem, NC, United States
| | - Ann Craddock
- Wake Forest University School of Medicine, Department of Biochemistry, Winston Salem, NC, United States
| | - Neeraj Kumar Sharma
- Wake Forest University School of Medicine, Department of Internal Medicine, Winston Salem, NC, United States
| | - Mary Elizabeth Comeau
- Wake Forest University School of Medicine, Department of Biostatistics and Data Sciences, Winston Salem, NC, United States
| | - Carl Langefeld
- Wake Forest University School of Medicine, Department of Biostatistics and Data Sciences, Winston Salem, NC, United States
| | - Gregory A Hawkins
- Wake Forest University School of Medicine, Department of Biochemistry, Winston Salem, NC, United States
| | - Richard Mott
- University College London, Department of Genetics, Evolution and Environment, Division of Biosciences, London, United Kingdom
| | - William Valdar
- University of North Carolina at Chapel Hill, Department of Genetics, Chapel Hill, NC, United States
| | - Leah C Solberg Woods
- Wake Forest University School of Medicine, Department of Internal Medicine, Winston Salem, NC, United States
| |
Collapse
|
14
|
Zhai R, Pan L, Yang Z, Li T, Ning Z, Pawitan Y, Wilson JF, Wu D, Shen X. Genetic and phenotypic links between obesity and extracellular vesicles. Hum Mol Genet 2022; 31:3643-3651. [PMID: 35357430 PMCID: PMC9616576 DOI: 10.1093/hmg/ddac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/12/2022] [Accepted: 03/17/2022] [Indexed: 11/14/2022] Open
Abstract
Obesity has a highly complex genetic architecture, making it difficult to understand the genetic mechanisms, despite the large number of discovered loci via genome-wide association studies (GWAS). Omics techniques have provided a better resolution to view this problem. As a proxy of cell-level biology, extracellular vesicles (EVs) are useful for studying cellular regulation of complex phenotypes such as obesity. Here, in a well-established Scottish cohort, we utilized a novel technology to detect surface proteins across millions of single EVs in each individual's plasma sample. Integrating the results with established obesity GWAS, we inferred 78 types of EVs carrying one or two of 12 surface proteins to be associated with adiposity-related traits such as waist circumference. We then verified that particular EVs' abundance is negatively correlated with body adiposity, while no association with lean body mass. We also revealed that genetic variants associated with protein-specific EVs capture 2-4-fold heritability enrichment for blood cholesterol levels. Our findings provide evidence that EVs with specific surface proteins have phenotypic and genetic links to obesity and blood lipids, respectively, guiding future EV biomarker research.
Collapse
Affiliation(s)
| | - Lu Pan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Zhijian Yang
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China,Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou 511458, China
| | - Ting Li
- Biostatistics Group, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China,Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou 511458, China
| | - Zheng Ning
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| | | | - Di Wu
- To whom correspondence should be addressed at: Greater Bay Area Institute of Precision Medicine (Guangzhou), 2nd Nanjiang Road, Nansha District, Guangzhou 511458, China. Tel: +86 1381153502; ; Vesicode AB, Nobels väg 16, Solna 17165, Sweden. Tel: +46 739191116;
| | - Xia Shen
- To whom correspondence should be addressed at: Greater Bay Area Institute of Precision Medicine (Guangzhou), 2nd Nanjiang Road, Nansha District, Guangzhou 511458, China. Tel: +86 1381153502; ; Vesicode AB, Nobels väg 16, Solna 17165, Sweden. Tel: +46 739191116;
| |
Collapse
|
15
|
Toyoda S, Shin J, Fukuhara A, Otsuki M, Shimomura I. Transforming growth factor β1 signaling links extracellular matrix remodeling to intracellular lipogenesis upon physiological feeding events. J Biol Chem 2022; 298:101748. [PMID: 35189145 PMCID: PMC8931428 DOI: 10.1016/j.jbc.2022.101748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue dynamically changes its mass in response to external nutritional status, which plays an important role in maintaining the lipid homeostasis. Physiologically, feeding events are associated with the expansion of adipose tissue, but little is known about the detailed molecular mechanisms of this expansion. Here, using comprehensive transcriptome analysis, we found that levels of transforming growth factor β1 (TGF-β1), a key regulator of extracellular matrix (ECM) remodeling, were increased in adipose tissue under feeding conditions and associated with the lipogenic pathway. In addition, TGF-β receptors are highly expressed in adipose tissue, and pharmacological inhibition of TGF-β1 reduced adipose tissue mass and caused ectopic lipid accumulation in the liver. This reduced fat mass was associated with decreased gene expression in ECM remodeling and lipogenesis. Furthermore, similar results were observed in the adipose tissue of SMAD family member 3 knockout mice or upon systemic TGF-β neutralization, with significant reductions in both ECM remodeling and lipogenesis-related genes. Mechanistically, we found that insulin-induced TGF-β1 and cell-autonomous action remodels the ECM of adipocytes, which controls the downstream focal adhesion kinase–AKT signaling cascades and enhances the lipogenic pathway. Of note, destruction of collagens or matrix metalloproteinase/a disintegrin and metalloprotease activities, critical components of ECM remodeling, blocked TGF-β1-mediated focal adhesion kinase–AKT signaling and the lipogenic pathway. Taken together, this study identifies a previously unknown lipogenic role of TGF-β1 by which adipocytes can expand to adapt to physiological feeding events.
Collapse
Affiliation(s)
- Shinichiro Toyoda
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jihoon Shin
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; Department of Diabetes Care Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Atsunori Fukuhara
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; Department of Adipose Management, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Michio Otsuki
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
16
|
Guo X, Sunil C, Qian G. Obesity and the Development of Lung Fibrosis. Front Pharmacol 2022; 12:812166. [PMID: 35082682 PMCID: PMC8784552 DOI: 10.3389/fphar.2021.812166] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Obesity is an epidemic worldwide and the obese people suffer from a range of respiratory complications including fibrotic changes in the lung. The influence of obesity on the lung is multi-factorial, which is related to both mechanical injury and various inflammatory mediators produced by excessive adipose tissues, and infiltrated immune cells. Adiposity causes increased production of inflammatory mediators, for example, cytokines, chemokines, and adipokines, both locally and in the systemic circulation, thereby rendering susceptibility to respiratory diseases, and altered responses. Lung fibrosis is closely related to chronic inflammation in the lung. Current data suggest a link between lung fibrosis and diet-induced obesity, although the mechanism remains incomplete understood. This review summarizes findings on the association of lung fibrosis with obesity, highlights the role of several critical inflammatory mediators (e.g., TNF-α, TGF-β, and MCP-1) in obesity related lung fibrosis and the implication of obesity in the outcomes of idiopathic pulmonary fibrosis patients.
Collapse
Affiliation(s)
- Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Christudas Sunil
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| |
Collapse
|
17
|
Kumari R, Irudayam MJ, Al Abdallah Q, Jones TL, Mims TS, Puchowicz MA, Pierre JF, Brown CW. SMAD2 and SMAD3 differentially regulate adiposity and the growth of subcutaneous white adipose tissue. FASEB J 2021; 35:e22018. [PMID: 34731499 DOI: 10.1096/fj.202101244r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/28/2021] [Accepted: 10/13/2021] [Indexed: 11/11/2022]
Abstract
Adipose tissue is the primary site of energy storage, playing important roles in health. While adipose research largely focuses on obesity, fat also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety and total energy homeostasis. SMAD2/3 proteins are downstream mediators of activin signaling, which regulate critical preadipocyte and mature adipocyte functions. Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity. The direct contributions of Smad2 and Smad3 in adipose tissues, however, are unknown. Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 on diet-induced adiposity using Smad2 or Smad3 "floxed" mice intercrossed with Adiponectin-Cre mice. Additionally, we examined visceral and subcutaneous preadipocyte differentiation efficiency in vitro. Almost all wild type subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A suppressed differentiation of preadipocytes from both depots. Smad2 conditional knockout (Smad2cKO) mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3 conditional knockout (Smad3cKO) male mice displayed a trend of reduced body weight on high-fat diet. On both diets, Smad3cKO mice displayed an adipose depot-selective phenotype, with a significant reduction in subcutaneous fat mass but not visceral fat mass. Our data suggest that Smad3 is an important contributor to the maintenance of subcutaneous white adipose tissue in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation.
Collapse
Affiliation(s)
- Roshan Kumari
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Maria Johnson Irudayam
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Qusai Al Abdallah
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tamekia L Jones
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Children's Foundation Research Institute, Memphis, Tennessee, USA
| | - Tahliyah S Mims
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Joseph F Pierre
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chester W Brown
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| |
Collapse
|
18
|
Role of Inflammatory Cytokines, Growth Factors and Adipokines in Adipogenesis and Insulin Resistance. Inflammation 2021; 45:31-44. [PMID: 34536157 PMCID: PMC8449520 DOI: 10.1007/s10753-021-01559-z] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 01/06/2023]
Abstract
Obesity, manifested by increased adiposity, represents a main cause of morbidity in the developed countries, causing increased risk of insulin resistance and type 2 diabetes mellitus. Recruitment of macrophages and activation of innate immunity represent the initial insult, which can be further exacerbated through secretion of chemokines and adipocytokines from activated macrophages and other cells within the adipose tissue. These events can impact adipogenesis, causing dysfunction of the adipose tissue and increased risk of insulin resistance. Various factors mediate adiposity and related insulin resistance including inflammatory and non-inflammatory factors such as pro and anti-inflammatory cytokines, adipokines and growth factors. In this review we will discuss the role of these factors in adipogenesis and development of insulin resistance and type 2 diabetes mellitus in the context of obesity. Understanding the molecular mechanisms that mediate adipogenesis and insulin resistance could help the development of novel therapeutic strategies for individuals at higher risk of insulin resistance and type 2 diabetes mellitus.
Collapse
|
19
|
Halbgebauer D, Roos J, Funcke JB, Neubauer H, Hamilton BS, Simon E, Amri EZ, Debatin KM, Wabitsch M, Fischer-Posovszky P, Tews D. Latent TGFβ-binding proteins regulate UCP1 expression and function via TGFβ2. Mol Metab 2021; 53:101336. [PMID: 34481123 PMCID: PMC8456047 DOI: 10.1016/j.molmet.2021.101336] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
Objective Activation of brown adipose tissue (BAT) in humans has been proposed as a new treatment approach for combating obesity and its associated diseases, as BAT participates in the regulation of energy homeostasis as well as glucose and lipid metabolism. Genetic contributors driving brown adipogenesis in humans have not been fully understood. Methods Profiling the gene expression of progenitor cells from subcutaneous and deep neck adipose tissue, we discovered new secreted factors with potential regulatory roles in white and brown adipogenesis. Among these, members of the latent transforming growth factor beta-binding protein (LTBP) family were highly expressed in brown compared to white adipocyte progenitor cells, suggesting that these proteins are capable of promoting brown adipogenesis. To investigate this potential, we used CRISPR/Cas9 to generate LTBP-deficient human preadipocytes. Results We demonstrate that LTBP2 and LTBP3 deficiency does not affect adipogenic differentiation, but diminishes UCP1 expression and function in the obtained mature adipocytes. We further show that these effects are dependent on TGFβ2 but not TGFβ1 signaling: TGFβ2 deficiency decreases adipocyte UCP1 expression, whereas TGFβ2 treatment increases it. The activity of the LTBP3–TGFβ2 axis that we delineate herein also significantly correlates with UCP1 expression in human white adipose tissue (WAT), suggesting an important role in regulating WAT browning as well. Conclusions These results provide evidence that LTBP3, via TGFβ2, plays an important role in promoting brown adipogenesis by modulating UCP1 expression and mitochondrial oxygen consumption. Inhibition of LTBP2 and LTBP3 reduces secretion of TGFβ2. Both knockout of LTBP2/3 or TGFβ2 inhibit UCP1 expression and mitochondrial respiration in human adipocytes. Expression of TGFβ2 correlates with UCP1 expression in human adipose tissue. Treatment with TGFβ2 rescues inhibition of UCP1 by LTBP knockout during adipogenesis.
Collapse
Affiliation(s)
- D Halbgebauer
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany; Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - J Roos
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - J B Funcke
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - H Neubauer
- Cardiometabolic Diseases Research, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - B S Hamilton
- Cardiometabolic Diseases Research, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - E Simon
- Global Computational Biology and Digital Sciences, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - E Z Amri
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - K M Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - M Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - P Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - D Tews
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany; Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
20
|
Bruno A, Di Sano C, Simon HU, Chanez P, Patti AM, Di Vincenzo S, Dino P, D'Esposito V, Formisano P, Beguinot F, Pace E. Leptin and TGF-β1 Downregulate PREP1 Expression in Human Adipose-Derived Mesenchymal Stem Cells and Mature Adipocytes. Front Cell Dev Biol 2021; 9:700481. [PMID: 34327205 PMCID: PMC8315375 DOI: 10.3389/fcell.2021.700481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
Adipose tissue is widely recognized as an extremely active endocrine organ producing adipokines as leptin that bridge metabolism and the immune system. Pre-B-cell leukemia homeobox (Pbx)-regulating protein-1 (PREP1) is a ubiquitous homeodomain transcription factor involved in the adipogenic differentiation and insulin-sensitivity processes. Leptin, as pleiotropic adipokine, and TGF-β, known to be expressed by primary pre-adipocytes [adipose-derived stem cells (ASCs)] and mature differentiated adipocytes, modulate inflammatory responses. We aimed to assess for the first time if leptin and TGF-β interfere with PREP1 expression in both ASCs and mature differentiated adipocytes. Human ASCs were isolated from subcutaneous adipose liposuction and, after expansion, fully differentiated to mature adipocytes. In both ASCs and adipocytes, leptin and TGF-β1 significantly decreased the expression of PREP1, alone and following concurrent Toll-like receptor 4 (TLR4) activation. Moreover, in adipocytes, but not in ASCs, leptin increased TLR4 and IL-33 expression, whereas TGF-β1 enhanced TLR4 and IL-6 expression. Taken together, we provide evidence for a direct regulation of PREP1 by leptin and TGF-β1 in ASCs and mature adipocytes. The effects of leptin and TGF-β1 on immune receptors and cytokines, however, are limited to mature adipocytes, suggesting that modulating immune responses depends on the differentiation of ASCs. Further studies are needed to fully understand the regulation of PREP1 expression and its potential for the development of new therapeutic approaches in obesity-related diseases.
Collapse
Affiliation(s)
- Andreina Bruno
- Institute for Biomedical Research and Innovation (IRIB), National Research Council, Palermo, Italy
| | - Caterina Di Sano
- Institute for Biomedical Research and Innovation (IRIB), National Research Council, Palermo, Italy
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.,Institute of Biochemistry, Medical School Brandenburg, Neuruppin, Germany.,Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.,Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Pascal Chanez
- Department of Respiratory Diseases CIC Nord INSERM, INRAE, C2VN, Aix Marseille University, Marseille, France
| | - Angelo Maria Patti
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Serena Di Vincenzo
- Institute for Biomedical Research and Innovation (IRIB), National Research Council, Palermo, Italy
| | - Paola Dino
- Institute for Biomedical Research and Innovation (IRIB), National Research Council, Palermo, Italy
| | - Vittoria D'Esposito
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Pietro Formisano
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Francesco Beguinot
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy.,Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| | - Elisabetta Pace
- Institute for Biomedical Research and Innovation (IRIB), National Research Council, Palermo, Italy
| |
Collapse
|
21
|
A novel podocyte protein, R3h domain containing-like, inhibits TGF-β-induced p38 MAPK and regulates the structure of podocytes and glomerular basement membrane. J Mol Med (Berl) 2021; 99:859-876. [PMID: 33620517 DOI: 10.1007/s00109-021-02050-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 01/14/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022]
Abstract
Not only in kidney glomerular physiological function but also glomerular pathology especially in diabetic condition, glomerular podocytes play pivotal roles. Therefore, it is important to increase our knowledge about the genes and proteins expressed in podocytes. Recently, we have identified a novel podocyte-expressed gene, R3h domain containing-like (R3hdml) and analyzed its function in vivo as well as in vitro. Transforming growth factor-β (TGF-β) signaling regulated the expression of R3hdml. And R3hdml inhibited p38 mitogen-activated protein kinase phosphorylation, which was induced by TGF-β, leading to the amelioration of podocyte apoptosis. Furthermore, a lack of R3hdml in mice significantly worsened glomerular function in streptozotocin (STZ)-induced diabetes, while overexpression of R3hdml ameliorated albuminuria in STZ-induced diabetes. Our results surmise that the functional analyses of R3hdml may lead to the development of novel therapeutic strategies for diabetic nephropathy in the future. KEY MESSAGES: • A novel podocyte expressed protein R3h domain containing-like was identified. • R3HDML inhibits podocyte apoptosis by inhibiting TGF-β-mediated p38 MAPK signaling. • Overexpression of R3HDML ameliorates albuminuria in STZ-induced diabetes mice. • R3HDML may prove to be a novel therapeutic strategy for diabetic nephropathy.
Collapse
|
22
|
Bantulà M, Roca-Ferrer J, Arismendi E, Picado C. Asthma and Obesity: Two Diseases on the Rise and Bridged by Inflammation. J Clin Med 2021; 10:jcm10020169. [PMID: 33418879 PMCID: PMC7825135 DOI: 10.3390/jcm10020169] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Asthma and obesity are two epidemics affecting the developed world. The relationship between obesity and both asthma and severe asthma appears to be weight-dependent, causal, partly genetic, and probably bidirectional. There are two distinct phenotypes: 1. Allergic asthma in children with obesity, which worsens a pre-existing asthma, and 2. An often non allergic, late-onset asthma developing as a consequence of obesity. In obesity, infiltration of adipose tissue by macrophages M1, together with an increased expression of multiple mediators that amplify and propagate inflammation, is considered as the culprit of obesity-related inflammation. Adipose tissue is an important source of adipokines, such as pro-inflammatory leptin, produced in excess in obesity, and adiponectin with anti-inflammatory effects with reduced synthesis. The inflammatory process also involves the synthesis of pro-inflammatory cytokines such as IL-1β, IL-6, TNFα, and TGFβ, which also contribute to asthma pathogenesis. In contrast, asthma pro-inflammatory cytokines such as IL-4, IL-5, IL-13, and IL-33 contribute to maintain the lean state. The resulting regulatory effects of the immunomodulatory pathways underlying both diseases have been hypothesized to be one of the mechanisms by which obesity increases asthma risk and severity. Reduction of weight by diet, exercise, or bariatric surgery reduces inflammatory activity and improves asthma and lung function.
Collapse
Affiliation(s)
- Marina Bantulà
- Department of Internal Medicine, Hospital Clinic, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.B.); (J.R.-F.); (E.A.)
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Jordi Roca-Ferrer
- Department of Internal Medicine, Hospital Clinic, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.B.); (J.R.-F.); (E.A.)
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
| | - Ebymar Arismendi
- Department of Internal Medicine, Hospital Clinic, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.B.); (J.R.-F.); (E.A.)
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
- Servei de Pneumologia, Hospital Clinic, 08036 Barcelona, Spain
| | - César Picado
- Department of Internal Medicine, Hospital Clinic, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.B.); (J.R.-F.); (E.A.)
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-227-5400
| |
Collapse
|
23
|
Alshargabi R, Shinjo T, Iwashita M, Yamashita A, Sano T, Nishimura Y, Hayashi M, Zeze T, Fukuda T, Sanui T, Nishimura F. SPOCK1 induces adipose tissue maturation: New insights into the function of SPOCK1 in metabolism. Biochem Biophys Res Commun 2020; 533:1076-1082. [PMID: 33012508 DOI: 10.1016/j.bbrc.2020.09.129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 11/24/2022]
Abstract
SPOCK1 is a calcium-binding matricellular proteoglycan that has been extensively studied in several cancer cells. Previously, we generated a mouse line overexpressing SPOCK1 (Spock1-Tg mouse) and showed that SPOCK1 might play an important role in drug-induced gingival overgrowth, indicating that it possesses physiological functions in non-cancer diseases as well. Although SPOCK1 was reported to be secreted from human adipocytes, its role in adipocyte physiology has not been addressed yet. In this study, SPOCK1 protein expression was confirmed in pancreas, adipose tissues, spleen, and liver of normal diet (ND)-fed mice. Interestingly, SPOCK1 was up-regulated in the pancreas and adipose tissues of the high-fat diet (HFD)-fed mice. Spock1-Tg mice fed with ND showed increased maturation in epididymal and inguinal adipose tissues. In addition, Spock1 overexpression strongly decreased expression of UCP-1 in adipose tissues, suggesting that SPOCK1 might regulate thermogenic function through suppression of UCP-1 expression. Finally, exogenous SPOCK1 treatment directly accelerated the differentiation of 3T3-L1 adipocytes, accompanied by the up-regulation of adipocyte differentiation-related gene expression. In conclusion, we demonstrated for the first time that SPOCK1 induced adipocyte differentiation via the up-regulation of adipogenesis-related genes.
Collapse
Affiliation(s)
- Rehab Alshargabi
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takanori Shinjo
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Misaki Iwashita
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akiko Yamashita
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomomi Sano
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Nishimura
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masato Hayashi
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tatsuro Zeze
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takao Fukuda
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Terukazu Sanui
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Fusanori Nishimura
- Section of Periodontology, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
24
|
Leung KL, Sanchita S, Pham CT, Davis BA, Okhovat M, Ding X, Dumesic P, Grogan TR, Williams KJ, Morselli M, Ma F, Carbone L, Li X, Pellegrini M, Dumesic DA, Chazenbalk GD. Dynamic changes in chromatin accessibility, altered adipogenic gene expression, and total versus de novo fatty acid synthesis in subcutaneous adipose stem cells of normal-weight polycystic ovary syndrome (PCOS) women during adipogenesis: evidence of cellular programming. Clin Epigenetics 2020; 12:181. [PMID: 33228780 PMCID: PMC7686698 DOI: 10.1186/s13148-020-00970-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
Background Normal-weight polycystic ovary syndrome (PCOS) women exhibit adipose resistance in vivo accompanied by enhanced subcutaneous (SC) abdominal adipose stem cell (ASC) development to adipocytes with accelerated lipid accumulation per cell in vitro. The present study examines chromatin accessibility, RNA expression and fatty acid (FA) synthesis during SC abdominal ASC differentiation into adipocytes in vitro of normal-weight PCOS versus age- and body mass index-matched normoandrogenic ovulatory (control) women to study epigenetic/genetic characteristics as well as functional alterations of PCOS and control ASCs during adipogenesis. Results SC abdominal ASCs from PCOS women versus controls exhibited dynamic chromatin accessibility during adipogenesis, from significantly less chromatin accessibility at day 0 to greater chromatin accessibility by day 12, with enrichment of binding motifs for transcription factors (TFs) of the AP-1 subfamily at days 0, 3, and 12. In PCOS versus control cells, expression of genes governing adipocyte differentiation (PPARγ, CEBPα, AGPAT2) and function (ADIPOQ, FABP4, LPL, PLIN1, SLC2A4) was increased two–sixfold at days 3, 7, and 12, while that involving Wnt signaling (FZD1, SFRP1, and WNT10B) was decreased. Differential gene expression in PCOS cells at these time points involved triacylglycerol synthesis, lipid oxidation, free fatty acid beta-oxidation, and oxidative phosphorylation of the TCA cycle, with TGFB1 as a significant upstream regulator. There was a broad correspondence between increased chromatin accessibility and increased RNA expression of those 12 genes involved in adipocyte differentiation and function, Wnt signaling, as well as genes involved in the triacylglycerol synthesis functional group at day 12 of adipogenesis. Total content and de novo synthesis of myristic (C14:0), palmitic (C16:0), palmitoleic (C16:1), and oleic (C18:1) acid increased from day 7 to day 12 in all cells, with total content and de novo synthesis of FAs significantly greater in PCOS than controls cells at day 12. Conclusions In normal-weight PCOS women, dynamic chromatin remodeling of SC abdominal ASCs during adipogenesis may enhance adipogenic gene expression as a programmed mechanism to promote greater fat storage.
Collapse
Affiliation(s)
- Karen L Leung
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Smriti Sanchita
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Catherine T Pham
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Brett A Davis
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Sciences University, Portland, OR, 97239, USA
| | - Mariam Okhovat
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Sciences University, Portland, OR, 97239, USA
| | - Xiangming Ding
- Technology Center for Genomics and Bioinformatics, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | | | - Tristan R Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Kevin J Williams
- UCLA Lipidomics Lab, Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Marco Morselli
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Feiyang Ma
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Lucia Carbone
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Sciences University, Portland, OR, 97239, USA.,Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, OR, 97239, USA.,Department of Medical Information and Clinical Epidemiology, Oregon Health and Sciences University, Portland, OR, 97239, USA.,Division of Genetics, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Xinmin Li
- Technology Center for Genomics and Bioinformatics, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
25
|
Gharanei S, Shabir K, Brown JE, Weickert MO, Barber TM, Kyrou I, Randeva HS. Regulatory microRNAs in Brown, Brite and White Adipose Tissue. Cells 2020; 9:cells9112489. [PMID: 33207733 PMCID: PMC7696849 DOI: 10.3390/cells9112489] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/02/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) constitute a class of short noncoding RNAs which regulate gene expression by targeting messenger RNA, inducing translational repression and messenger RNA degradation. This regulation of gene expression by miRNAs in adipose tissue (AT) can impact on the regulation of metabolism and energy homeostasis, particularly considering the different types of adipocytes which exist in mammals, i.e., white adipocytes (white AT; WAT), brown adipocytes (brown AT; BAT), and inducible brown adipocytes in WAT (beige or brite or brown-in-white adipocytes). Indeed, an increasing number of miRNAs has been identified to regulate key signaling pathways of adipogenesis in BAT, brite AT, and WAT by acting on transcription factors that promote or inhibit adipocyte differentiation. For example, MiR-328, MiR-378, MiR-30b/c, MiR-455, MiR-32, and MiR-193b-365 activate brown adipogenesis, whereas MiR-34a, MiR-133, MiR-155, and MiR-27b are brown adipogenesis inhibitors. Given that WAT mainly stores energy as lipids, whilst BAT mainly dissipates energy as heat, clarifying the effects of miRNAs in different types of AT has recently attracted significant research interest, aiming to also develop novel miRNA-based therapies against obesity, diabetes, and other obesity-related diseases. Therefore, this review presents an up-to-date comprehensive overview of the role of key regulatory miRNAs in BAT, brite AT, and WAT.
Collapse
Affiliation(s)
- Seley Gharanei
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Kiran Shabir
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
| | - James E. Brown
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre of Applied Biological & Exercise Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
- Correspondence:
| |
Collapse
|
26
|
Roh HC, Kumari M, Taleb S, Tenen D, Jacobs C, Lyubetskaya A, Tsai LTY, Rosen ED. Adipocytes fail to maintain cellular identity during obesity due to reduced PPARγ activity and elevated TGFβ-SMAD signaling. Mol Metab 2020; 42:101086. [PMID: 32992037 PMCID: PMC7559520 DOI: 10.1016/j.molmet.2020.101086] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022] Open
Abstract
Objective Obesity due to overnutrition causes adipose tissue dysfunction, which is a critical pathological step on the road to type 2 diabetes (T2D) and other metabolic disorders. In this study, we conducted an unbiased investigation into the fundamental molecular mechanisms by which adipocytes transition to an unhealthy state during obesity. Methods We used nuclear tagging and translating ribosome affinity purification (NuTRAP) reporter mice crossed with Adipoq-Cre mice to determine adipocyte-specific 1) transcriptional profiles (RNA-seq), 2) promoter and enhancer activity (H3K27ac ChIP-seq), 3) and PPARγ cistrome (ChIP-seq) profiles in mice fed chow or a high-fat diet (HFD) for 10 weeks. We also assessed the impact of the PPARγ agonist rosiglitazone (Rosi) on gene expression and cellular state of adipocytes from the HFD-fed mice. We integrated these data to determine the transcription factors underlying adipocyte responses to HFD and conducted functional studies using shRNA-mediated loss-of-function approaches in 3T3-L1 adipocytes. Results Adipocytes from the HFD-fed mice exhibited reduced expression of adipocyte markers and metabolic genes and enhanced expression of myofibroblast marker genes involved in cytoskeletal organization, accompanied by the formation of actin filament structures within the cell. PPARγ binding was globally reduced in adipocytes after HFD feeding, and Rosi restored the molecular and cellular phenotypes of adipocytes associated with HFD feeding. We identified the TGFβ1 effector protein SMAD to be enriched at HFD-induced promoters and enhancers and associated with myofibroblast signature genes. TGFβ1 treatment of mature 3T3-L1 adipocytes induced gene expression and cellular changes similar to those seen after HFD in vivo, and knockdown of Smad3 blunted the effects of TGFβ1. Conclusions Our data demonstrate that adipocytes fail to maintain cellular identity after HFD feeding, acquiring characteristics of a myofibroblast-like cell type through reduced PPARγ activity and elevated TGFβ-SMAD signaling. This cellular identity crisis may be a fundamental mechanism that drives functional decline of adipose tissues during obesity. Adipocytes after HFD intake exhibit defects in cellular identity maintenance. Adipocytes develop actin filament networks in obesity. Altered PPARγ activity mediates defective adipocyte identity phenotypes. TGFβ-SMAD pathways promote HFD-induced aberrant phenotype of adipocytes.
Collapse
Affiliation(s)
- Hyun Cheol Roh
- Division of Endocrinology, Diabetes and Obesity, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA; Broad Institute, Cambridge, MA, 02142, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Manju Kumari
- Division of Endocrinology, Diabetes and Obesity, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Solaema Taleb
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Danielle Tenen
- Division of Endocrinology, Diabetes and Obesity, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA; Broad Institute, Cambridge, MA, 02142, USA
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes and Obesity, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA; Broad Institute, Cambridge, MA, 02142, USA
| | - Anna Lyubetskaya
- Division of Endocrinology, Diabetes and Obesity, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA; Broad Institute, Cambridge, MA, 02142, USA
| | - Linus T-Y Tsai
- Division of Endocrinology, Diabetes and Obesity, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA; Broad Institute, Cambridge, MA, 02142, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes and Obesity, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA; Broad Institute, Cambridge, MA, 02142, USA.
| |
Collapse
|
27
|
Zhang Z, Meng Y, Gao F, Xiao Y, Zheng Y, Wang HQ, Gao Y, Jiang H, Yuan B, Zhang JB. TGF-β1-Mediated FDNCR1 Regulates Porcine Preadipocyte Differentiation via the TGF-β Signaling Pathway. Animals (Basel) 2020; 10:ani10081399. [PMID: 32796679 PMCID: PMC7459525 DOI: 10.3390/ani10081399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Fat differentiation affects lipid deposition and is a complex metabolic process. It has been previously reported that multiple transcription factors regulate adipocyte formation. With the in-depth study of epigenetics, in recent years it has been reported that long noncoding RNA (lncRNA) can effectively affect the formation of lipid droplets and thus regulate fat deposition. lncRNA can regulate cell function through a variety of mechanisms, the most studied is the mechanism of action of lncRNA as a miRNA molecular sponge. The purpose of this article is to explore the role of transforming growth factor-beta (TGF-β1) mediated lncRNA in the formation of porcine adipocytes, from the perspective of lncRNA to reveal the effect of TGF-β1 on the differentiation of porcine adipocytes, and provide a new way to improve the quality of pork. Abstract Adipocyte differentiation and lipid metabolism have important regulatory effects on the quality of meat from livestock. A variety of transcription factors regulate preadipocyte differentiation. Several studies have revealed that transforming growth factor-beta (TGF-β1) may play a key role in epithelial–mesenchymal transition (EMT); however, little is known about the effects of TGF-β1 treatment on porcine preadipocytes. To explore the role of TGF-β1 in porcine adipocyte differentiation, porcine preadipocytes were treated with 10 ng/mL TGF-β1, and two libraries were constructed for RNA-seq. We chose an abundant and differentially expressed long noncoding RNA (lncRNA), which we named fat deposition-associated long noncoding RNA1 (FDNCR1), for further study. RT-qPCR was used to detect mRNA levels of genes related to adipocyte differentiation. Triglyceride assay kits were used to detect lipid droplet deposition. TGF-β1 significantly suppressed porcine preadipocyte differentiation. We identified 8158 lncRNAs in total and 39 differentially expressed lncRNAs. After transfection with FDNCR1 siRNA, the mRNA expression of aP2, C/EBPα, and PPARγ and triglyceride levels significantly increased. Transfection with FDNCR1 siRNA significantly decreased protein levels of p-Smad2/Smad2 and p-Smad3/Smad3. These results demonstrate that FDNCR1 suppresses porcine preadipocyte differentiation via the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Yu Meng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
| | - Fei Gao
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
| | - Yue Xiao
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
| | - Yi Zheng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
| | - Hao-Qi Wang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
| | - Yan Gao
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
| | - Hao Jiang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
| | - Bao Yuan
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
- Correspondence: (B.Y.); (J.-B.Z.); Tel.: +86-431-8783-6536 (B.Y.); +86-431-8783-6551 (J.-B.Z.)
| | - Jia-Bao Zhang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin 130062, China; (Z.Z.); (Y.M.); (F.G.); (Y.X.); (Y.Z.); (H.-Q.W.); (Y.G.); (H.J.)
- Correspondence: (B.Y.); (J.-B.Z.); Tel.: +86-431-8783-6536 (B.Y.); +86-431-8783-6551 (J.-B.Z.)
| |
Collapse
|
28
|
Nasias D, Dalakoura-Karagkouni K, Vassou D, Papagiannakis G, Papadaki A, Kardassis D. Transcriptome analysis of the adipose tissue in a mouse model of metabolic syndrome identifies gene signatures related to disease pathogenesis. Genomics 2020; 112:4053-4062. [PMID: 32652102 DOI: 10.1016/j.ygeno.2020.06.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/12/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
Abstract
The white adipose tissue (WAT) contributes to the metabolic imbalance observed in obesity and the metabolic syndrome (MetS) by mechanisms that are poorly understood. The aim of this study was to monitor changes in the transcriptome of epididymal WAT during the development of MetS. ApoE3L.CETP mice were fed a high fat (HFD) or a low-fat (LFD) diet for different time periods. Adipose RNA was analyzed by microarrays. We found an increasing number of differentially expressed transcripts during MetS development. In mice with MetS, 1396 transcripts were differentially expressed including transcripts related to immune/inflammatory responses and extracellular matrix enzymes, suggesting significant inflammation and tissue remodeling. The top list of pathways included focal adhesion, chemokine, B and T cell receptor and MAPK signaling. The data identify for the first time adipose gene signatures in apoE3L.CETP mice with diet-induced MetS and might open new avenues for investigation of potential biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Dimitris Nasias
- Laboratory of Biochemistry, Division of Basic Sciences, University of Crete Medical School, Heraklion 71003, Greece; Gene Regulation and Epigenetics group, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 70013, Greece
| | - Katerina Dalakoura-Karagkouni
- Laboratory of Biochemistry, Division of Basic Sciences, University of Crete Medical School, Heraklion 71003, Greece; Gene Regulation and Epigenetics group, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 70013, Greece
| | - Despoina Vassou
- Genomics Facility, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 70013, Greece
| | - Giorgos Papagiannakis
- Genomics Facility, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 70013, Greece
| | - Ariadni Papadaki
- Laboratory of Biochemistry, Division of Basic Sciences, University of Crete Medical School, Heraklion 71003, Greece
| | - Dimitris Kardassis
- Laboratory of Biochemistry, Division of Basic Sciences, University of Crete Medical School, Heraklion 71003, Greece; Gene Regulation and Epigenetics group, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 70013, Greece.
| |
Collapse
|
29
|
Function and characterization of the promoter region of perilipin 1 (PLIN1): Roles of E2F1, PLAG1, C/EBPβ, and SMAD3 in bovine adipocytes. Genomics 2020; 112:2400-2409. [DOI: 10.1016/j.ygeno.2020.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/01/2020] [Accepted: 01/21/2020] [Indexed: 12/23/2022]
|
30
|
Khan R, Raza SHA, Schreurs N, Xiaoyu W, Hongbao W, Ullah I, Rahman A, Suhail SM, Khan S, Linsen Z. Bioinformatics analysis and transcriptional regulation of TORC1 gene through transcription factors NRF1 and Smad3 in bovine preadipocytes. Genomics 2020; 112:1575-1587. [DOI: 10.1016/j.ygeno.2019.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/23/2019] [Accepted: 09/11/2019] [Indexed: 12/25/2022]
|
31
|
Pichiah PBT, Sankarganesh D, Arunachalam S, Achiraman S. Adipose-Derived Molecules-Untouched Horizons in Alzheimer's Disease Biology. Front Aging Neurosci 2020; 12:17. [PMID: 32116650 PMCID: PMC7032035 DOI: 10.3389/fnagi.2020.00017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/20/2020] [Indexed: 12/18/2022] Open
Abstract
The global incidence of Alzheimer's disease (AD) is on the rise with the increase in obesity and metabolic disease epidemic. Obesity is co-morbid with the increase in mass of adipose tissue, which secretes numerous molecules that are biologically important. Obesity and its associated conditions are perhaps involved in the causative pathway of AD. Immunologically important cytokines such as IL-1β, IL-10, and IL-18, which are released by adipose tissue, are also found to be associated with AD. Besides, the expression of IL-6, IFNγ, and TNF alpha are also associated with AD. Ang-I and Ang-II are found to mediate the progression of AD. Complement factors B, C4b, and H are differentially expressed in AD. Overall, several adipocyte-derived cytokines are found to be dysregulated in AD, and their role in AD remains to be studied. The induction of autophagy is a very promising strategy in the treatment of AD. A variety of adipose-derived molecules have been shown to modulate autophagy. However, very little literature is available on the role of adipose-derived molecules in inducing autophagy in microglial cells of AD. Understanding the role of adipose-derived molecules in the development of AD, especially in the induction of autophagy, would open up new avenues in devising strategies for the treatment of AD.
Collapse
Affiliation(s)
| | - Devaraj Sankarganesh
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, India
- Department of Microbial Biotechnology, Bharathiar University, Coimbatore, India
| | - Sankarganesh Arunachalam
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, India
| | - Shanmugam Achiraman
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, India
| |
Collapse
|
32
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
33
|
Sakamoto K, Furuichi Y, Yamamoto M, Takahashi M, Akimoto Y, Ishikawa T, Shimizu T, Fujimoto M, Takada-Watanabe A, Hayashi A, Mita Y, Manabe Y, Fujii NL, Ishibashi R, Maezawa Y, Betsholtz C, Yokote K, Takemoto M. R3hdml regulates satellite cell proliferation and differentiation. EMBO Rep 2019; 20:e47957. [PMID: 31524320 DOI: 10.15252/embr.201947957] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/06/2019] [Accepted: 08/16/2019] [Indexed: 12/29/2022] Open
Abstract
In this study, we identified a previously uncharacterized skeletal satellite cell-secreted protein, R3h domain containing-like (R3hdml). Expression of R3hdml increases during skeletal muscle development and differentiation in mice. Body weight and skeletal muscle mass of R3hdml knockout (KO) mice are lower compared to control mice. Expression levels of cell cycle-related markers, phosphorylation of Akt, and expression of insulin-like growth factor within the skeletal muscle are reduced in R3hdml KO mice compared to control mice. Expression of R3hdml increases during muscle regeneration in response to cardiotoxin (CTX)-induced muscle injury. Recovery of handgrip strength after CTX injection was significantly impaired in R3hdml KO mice, which is rescued by R3hdml. Our results indicate that R3hdml is required for skeletal muscle development, regeneration, and, in particular, satellite cell proliferation and differentiation.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yasuro Furuichi
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Masashi Yamamoto
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Megumi Takahashi
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Yoshihiro Akimoto
- Department of Anatomy, Kyorin University School of Medicine, Tokyo, Japan
| | - Takahiro Ishikawa
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan.,Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba, Japan
| | - Takahiko Shimizu
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan.,Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Masanori Fujimoto
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Aki Takada-Watanabe
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Aiko Hayashi
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshitaka Mita
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Yasuko Manabe
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Nobuharu L Fujii
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Ryoichi Ishibashi
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan.,Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba, Japan
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Cancer and Vascular Biology, Uppsala Universitet, Uppsala, Sweden.,Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, Novum, Huddinge, Sweden
| | - Koutaro Yokote
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan.,Division of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba, Japan
| | - Minoru Takemoto
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Diabetes, Metabolism and Endocrinology, School of Medicine, International University of Health and Welfare, Narita, Chiba, Japan
| |
Collapse
|
34
|
Zhang Z, Wang W, Liu JB, Wang Y, Hao JD, Huang YJ, Gao Y, Jiang H, Yuan B, Zhang JB. ssc-miR-204 regulates porcine preadipocyte differentiation and apoptosis by targeting TGFBR1 and TGFBR2. J Cell Biochem 2019; 121:609-620. [PMID: 31353638 DOI: 10.1002/jcb.29306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) take part in a variety of biological processes by regulating target genes. Transforming growth factor β receptor 1 (TGFBR1) and TGFBR2 are crucial members of the TGF-β family and are serine/threonine kinase receptors. The aim of this study was to explore the functions of ssc-miR-204 in porcine preadipocyte differentiation and apoptosis with regard to the TGFβ/Smad pathway. We identified miRNAs predicted to target TGFBR1 and TGFBR2 using a database and selected ssc-miR-204 as a candidate miRNA. ssc-miR-204 overexpression dramatically reduced the levels of TGFBR1 and TGFBR2. However, after transfection with ssc-miR-204 inhibitor, TGFBR1 and TGFBR2 levels were dramatically increased. ssc-miR-204 overexpression dramatically promoted porcine preadipocyte differentiation and apoptosis. After transfection with ssc-miR-204 inhibitor, porcine preadipocyte differentiation and apoptosis were dramatically inhibited. After transfection with ssc-miR-204 mimics, Smad2, Smad3, Smad4, p-Smad2, and p-Smad3 protein levels significantly decreased, and adipogenesis was regulated by inhibiting the TGF-β/Smad3 signaling pathway. Taken together, these results verified that ssc-miR-204 regulates porcine preadipocyte differentiation and apoptosis by targeting TGFBR1 and TGFBR2.
Collapse
Affiliation(s)
- Zhe Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Wei Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Jian-Bo Liu
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Ying Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Jin-Dong Hao
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Yi-Jie Huang
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Yan Gao
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Hao Jiang
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Bao Yuan
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | - Jia-Bao Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
35
|
Tan J, Wang Y, Wang S, Wu S, Yuan Z, Zhu X. Label-free quantitative proteomics identifies transforming growth factor β1 (TGF-β1) as an inhibitor of adipogenic transformation in OP9-DL1 cells and primary thymic stromal cells. Cell Biosci 2019; 9:48. [PMID: 31249661 PMCID: PMC6570845 DOI: 10.1186/s13578-019-0311-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/08/2019] [Indexed: 11/21/2022] Open
Abstract
Background Adipocyte accumulation is a predominant feature of age-related thymic involution, but the mechanisms responsible for thymic adipogenesis remain to be elucidated. The aim of this study was to identify key regulators in thymic adipogenesis. We used rosiglitazone, a potent peroxisome proliferator-activated receptor γ (PPARγ) agonist, to induce adipogenic differentiation of OP9-DL1 cells, and investigated the differentially expressed proteins during adipogenic differentiation by using label-free quantitative proteomics. Furthermore, the effects of transforming growth factor β1 (TGF-β1) on rosiglitazone-induced adipogenic differentiation of OP9-DL1 cells as well as the underlying mechanisms were also investigated. Results Proteomic analysis identified 139 proteins differed significantly in rosiglitazone-treated cells compared with dimethyl sulphoxide (DMSO)-treated cells. Rosiglitazone-induced adipogenic differentiation was inhibited by TGF-β1 treatment in OP9-DL1 cells and primary thymic stromal cells. Real-time PCR analysis showed significant increases in PPARγ and fatty acid binding protein 4 mRNA levels in rosiglitazone-treated cells, which were inhibited by TGF-β1 treatment. TGF-β1 down-regulated PPARγ expression at both mRNA and protein levels in OP9-DL1 cells. Chromatin immunoprecipitation analysis demonstrated that TGF-β1 enhanced the binding of Smad2/3 and histone deacetylase 1, but reduced the binding of H3K14ac to the promoter of PPARγ gene. TGF-β1 partially reversed the inhibitory effects of rosiglitazone on the expression of Axin2 and β-catenin protein levels. TGF-β1 inhibited rosiglitazone-induced adipogenic transformation in OP9-DL1 cells by down-regulation of PPARγ and activation of the canonical Wnt/β-catenin signaling pathway. Conclusion Taken together, activation of TGF-β pathway may serve as a useful strategy to prevent thymic adiposity in age-related thymic involution. Electronic supplementary material The online version of this article (10.1186/s13578-019-0311-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianxin Tan
- 1State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004 People's Republic of China
| | - Yajun Wang
- 2Research Center, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People's Republic of China
| | - Siliang Wang
- 3Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Simeng Wu
- 4Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Zhe Yuan
- 4Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Xike Zhu
- 2Research Center, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People's Republic of China
| |
Collapse
|
36
|
Dumesic DA, Phan JD, Leung KL, Grogan TR, Ding X, Li X, Hoyos LR, Abbott DH, Chazenbalk GD. Adipose Insulin Resistance in Normal-Weight Women With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2019; 104:2171-2183. [PMID: 30649347 PMCID: PMC6482023 DOI: 10.1210/jc.2018-02086] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022]
Abstract
CONTEXT Normal-weight women with polycystic ovary syndrome (PCOS) may have adipose tissue insulin resistance (adipose-IR). OBJECTIVE To examine whether adipose-IR and subcutaneous (SC) abdominal adipose stem cell (ASC) gene expression are altered in normal-weight women with PCOS and correlated with hyperandrogenemia and/or whole-body IR. DESIGN Prospective cohort study. SETTING Academic medical center. PATIENTS Ten normal-weight women with PCOS and 18 control subjects matched for age and body mass index. INTERVENTION(S) Women underwent circulating hormone and metabolic measurements, IV glucose tolerance testing, total-body dual-energy x-ray absorptiometry, and SC abdominal fat biopsy. MAIN OUTCOME MEASURE(S) Adipose-IR (fasting insulin × total fatty acid levels) and SC abdominal ASC gene expression were compared between groups and correlated with clinical outcomes. RESULTS Adipose-IR was greater in women with PCOS than in control subjects (P < 0.01), with 29 pmol/L × mmol/L providing 94% specificity and 80% sensitivity in discriminating the two groups (P < 0.001). Adipose-IR positively correlated with serum androgen and log of fasting triglyceride (TG) levels, percentage of small adipocytes (P < 0.01, all correlations), and acute insulin response to glucose (P < 0.05); and negatively correlated with insulin sensitivity (Si; P < 0.025) and serum adiponectin levels (P < 0.05). Adjusting for serum androgens, adipose-IR correlations with Si and log TG levels remained significant. ASC genes were differentially expressed by the two groups. Expression of functionally critical genes was associated with serum testosterone and/or fasting insulin levels. CONCLUSION Normal-weight women with PCOS have increased adipose-IR and altered ASC gene expression related to hyperandrogenism and IR.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, California
- Correspondence and Reprint Requests: Daniel A. Dumesic, MD, Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, 10833 Le Conte Avenue, Room 22-178 CHS, Los Angeles, California 90095. E-mail:
| | - Julia D Phan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, California
| | - Karen L Leung
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, California
| | - Tristan R Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine at University of California, Los Angeles, California
| | - Xiangmiang Ding
- Technology Center for Genomics and Bioinformatics, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, California
| | - Xinmin Li
- Technology Center for Genomics and Bioinformatics, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, California
| | - Luis R Hoyos
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, California
| | - David H Abbott
- Department of Obstetrics and Gynecology and Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, California
| |
Collapse
|
37
|
Zhou S, Chen S, Jiang Q, Pei M. Determinants of stem cell lineage differentiation toward chondrogenesis versus adipogenesis. Cell Mol Life Sci 2019; 76:1653-1680. [PMID: 30689010 PMCID: PMC6456412 DOI: 10.1007/s00018-019-03017-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/10/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Adult stem cells, also termed as somatic stem cells, are undifferentiated cells, detected among differentiated cells in a tissue or an organ. Adult stem cells can differentiate toward lineage specific cell types of the tissue or organ in which they reside. They also have the ability to differentiate into mature cells of mesenchymal tissues, such as cartilage, fat and bone. Despite the fact that the balance has been comprehensively scrutinized between adipogenesis and osteogenesis and between chondrogenesis and osteogenesis, few reviews discuss the relationship between chondrogenesis and adipogenesis. In this review, the developmental and transcriptional crosstalk of chondrogenic and adipogenic lineages are briefly explored, followed by elucidation of signaling pathways and external factors guiding lineage determination between chondrogenic and adipogenic differentiation. An in-depth understanding of overlap and discrepancy between these two mesenchymal tissues in lineage differentiation would benefit regeneration of high-quality cartilage tissues and adipose tissues for clinical applications.
Collapse
Affiliation(s)
- Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, 610083, Sichuan, People's Republic of China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, School of Medicine, Drum Tower Hospital, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- Robert C. Byrd Health Sciences Center, WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
38
|
Zhang Z, Gao Y, Xu MQ, Wang CJ, Fu XH, Liu JB, Han DX, Jiang H, Yuan B, Zhang JB. miR-181a regulate porcine preadipocyte differentiation by targeting TGFBR1. Gene 2019; 681:45-51. [DOI: 10.1016/j.gene.2018.09.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022]
|
39
|
El-Tahlawi S, Ezzat Mohammad N, Mohamed El-Amir A, Sayed Mohamed H. Survivin and insulin-like growth factor-I: potential role in the pathogenesis of acne and post-acne scar. Scars Burn Heal 2019; 5:2059513118818031. [PMID: 30675395 PMCID: PMC6330724 DOI: 10.1177/2059513118818031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDS/OBJECTIVES Acne is an inflammatory disease of the pilosebaceous unit (PSU). The over-expression of survivin and insulin-like growth factor (IGF)-I in some fibrotic disorders suggests a possible implication in the pathogenesis of acne and or post-acne scar. We aimed to evaluate their potential role in pathogenesis in acne and post-acne scar. METHODS Serum survivin and IGF-I levels were estimated in 30 patients with acne and post-acne scar compared to 30 controls. RESULTS There was a statistically significant difference in survivin and IGF-I levels between controls and patients (P < 0.05). However, there was no linear correlation between survivin and IGF-I. CONCLUSIONS Survivin and IGF-I could have a possible role in the pathogenesis of active acne and in post-inflammatory acne scar.
Collapse
|
40
|
Engineered systems to study the synergistic signaling between integrin-mediated mechanotransduction and growth factors (Review). Biointerphases 2018; 13:06D302. [DOI: 10.1116/1.5045231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
41
|
CCN5 knockout mice exhibit lipotoxic cardiomyopathy with mild obesity and diabetes. PLoS One 2018; 13:e0207228. [PMID: 30485307 PMCID: PMC6261567 DOI: 10.1371/journal.pone.0207228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 10/26/2018] [Indexed: 01/01/2023] Open
Abstract
Obesity is associated with various human disorders, such as type 2 diabetes, cardiovascular diseases, hypertension, and cancers. In this study, we observed that knockout (KO) of CCN5, which encodes a matricellular protein, caused mild obesity in mice. The CCN5 KO mice also exhibited mild diabetes characterized by high fasting glucose levels and impaired insulin and glucose tolerances. Cardiac hypertrophy, ectopic lipid accumulation, and impaired lipid metabolism in hearts were observed in the CCN5 KO mice, as determined using histology, quantitative RT-PCR, and western blotting. Fibrosis was significantly greater in hearts from the CCN5 KO mice both in interstitial and perivascular regions, which was accompanied by higher expression of pro-fibrotic and pro-inflammatory genes. Both systolic and diastolic functions were significantly impaired in hearts from the CCN5 KO mice, as assessed using echocardiography. Taken together, these results indicate that CCN5 KO leads to lipotoxic cardiomyopathy with mild obesity and diabetes in mice.
Collapse
|
42
|
Strycharz J, Świderska E, Wróblewski A, Podolska M, Czarny P, Szemraj J, Balcerczyk A, Drzewoski J, Kasznicki J, Śliwińska A. Hyperglycemia Affects miRNAs Expression Pattern during Adipogenesis of Human Visceral Adipocytes-Is Memorization Involved? Nutrients 2018; 10:E1774. [PMID: 30445791 PMCID: PMC6266776 DOI: 10.3390/nu10111774] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 01/22/2023] Open
Abstract
microRNAs are increasingly analyzed in adipogenesis, whose deregulation, especially visceral, contributes to the development of diabetes. Hyperglycemia is known to affect cells while occurring acutely and chronically. Therefore, we aimed to evaluate the effect of hyperglycemia on human visceral pre/adipocytes from the perspective of microRNAs. The relative expression of 78 microRNAs was determined by TaqMan Low Density Arrays at three stages of HPA-v adipogenesis conducted under normoglycemia, chronic, and intermittent hyperglycemia (30 mM). Hierarchical clustering/Pearson correlation revealed the relationship between various microRNAs' expression profiles, while functional analysis identified the genes and signaling pathways regulated by differentially expressed microRNAs. Hyperglycemia affected microRNAs' expression patterns during adipogenesis, and at the stage of pre-adipocytes, differentiated and matured adipocytes compared to normoglycemia. Interestingly, the changes that were evoked upon hyperglycemic exposure during one adipogenesis stage resembled those observed upon chronic hyperglycemia. At least 15 microRNAs were modulated during normoglycemic and/or hyperglycemic adipogenesis and/or upon intermittent/chronic hyperglycemia. Bioinformatics analysis revealed the involvement of these microRNAs in cell cycles, lipid metabolism, ECM⁻receptor interaction, oxidative stress, signaling of insulin, MAPK, TGF-β, p53, and more. The obtained data suggests that visceral pre/adipocytes exposed to chronic/intermittent hyperglycemia develop a microRNAs' expression pattern, which may contribute to further visceral dysfunction, the progression of diabetic phenotype, and diabetic complications possibly involving "epi"-memory.
Collapse
Affiliation(s)
- Justyna Strycharz
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland.
| | - Ewa Świderska
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland.
| | - Adam Wróblewski
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland.
| | - Marta Podolska
- Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, 92-213 Lodz, Poland.
| | - Piotr Czarny
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland.
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland.
| | - Aneta Balcerczyk
- Department of Molecular Biophysics, University of Lodz, 90-236 Lodz, Poland.
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 92-213 Lodz, Poland.
| | - Jacek Kasznicki
- Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, 92-213 Lodz, Poland.
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213, 92-213 Lodz, Poland.
| |
Collapse
|
43
|
Wang X, Xu S, Chen L, Shen D, Cao Y, Tang R, Wang X, Ji C, Li Y, Cui X, Guo X. Profiling Analysis Reveals the Potential Contribution of Peptides to Human Adipocyte Differentiation. Proteomics Clin Appl 2018; 12:e1700172. [PMID: 30009563 DOI: 10.1002/prca.201700172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 05/15/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Peptide drugs provide promising regimes in anti-obesity treatment. In order to identify potential bioactive peptides involved in adipogenesis. EXPERIMENTAL DESIGN The intracellular peptides between preadipocytes and adipocytes are compared by liquid chromatography/mass spectrometry. The underlying biological function of the identified peptides are evaluated by gene ontology (GO) and pathway analysis of their precursors. To find more potential bioactive peptides, 50 peptide sequences are identified located in the functional domains with the use of the SMART and UniProt databases. Finally, the Open Targets Platform database is used to investigate the precursors related to metabolic diseases. RESULTS A total of 181 downregulated peptides and 89 upregulated peptides after differentiation (fold change > 1.5 and p-value < 0.05) are identified. The GO and pathway analysis indicate that these differentially expressed peptides play a role in adipogenesis. A total of 10 putative peptides 6 to 26 amino acids in length are identified that might have bioactive effects in adipogenesis and metabolic diseases. CONCLUSIONS AND CLINICAL RELEVANCE On one hand, present preliminary research provides a better understanding of the intracellular peptides during adipocyte differentiation. On the other hand, it lays a foundation for discovering new peptide drugs in anti-obesity treatment.
Collapse
Affiliation(s)
- Xingyun Wang
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Siliang Xu
- Center of Clinical Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Ling Chen
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Dan Shen
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Ranran Tang
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xing Wang
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Yun Li
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xianwei Cui
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xirong Guo
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| |
Collapse
|
44
|
Chu SH, Loucks EB, Kelsey KT, Gilman SE, Agha G, Eaton CB, Buka SL, Huang YT. Sex-specific epigenetic mediators between early life social disadvantage and adulthood BMI. Epigenomics 2018; 10:707-722. [PMID: 29888956 DOI: 10.2217/epi-2017-0146] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM The objective of this study was to identify potential epigenetic mediating pathways linking early life social disadvantage (ELSD) to adulthood BMI. METHODS Sex-specific epigenome-wide two-stage mediation analyses were conducted in blood and adipose tissue, and mediation estimates were obtained using cross-product mediation analysis. Pathway analyses were conducted using GREAT software (Bejerano Lab, CA, USA). RESULTS Candidate mediation CpG sites were identified in adipose tissue, but not blood, and were sex-specific. Significant mediation sites in females included CpG loci in genes: PKHG1, BCAR3, ADAM5P, PIEZO1, FGFRL1, FASN and DPP9, among others. Pathway analyses revealed evidence of enrichment for processes associated with TFG-β signaling and immunologic signatures. In males, significant mediation loci included sites in MAP3K5 and RPTOR, which have previously been associated with adipogenesis, inflammation and insulin resistance. CONCLUSION Our findings provide supportive evidence for the mediating role of epigenetic mechanisms in the effect of early life social disadvantage on adulthood BMI.
Collapse
Affiliation(s)
- Su H Chu
- Department of Epidemiology, Brown School of Public Health, Providence, RI, 02912, USA.,Channing Division of Network Medicine, Brigham & Women's Hospital & Harvard Medical School, Boston, MA, 02115, USA
| | - Eric B Loucks
- Department of Epidemiology, Brown School of Public Health, Providence, RI, 02912, USA
| | - Karl T Kelsey
- Department of Epidemiology, Brown School of Public Health, Providence, RI, 02912, USA.,Department of Pathology & Laboratory Medicine, Brown University Warren Alpert Medical School, Providence, RI, 02912, USA
| | - Stephen E Gilman
- Health Behavior Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health & Human Development, Bethesda, MD, 20892, USA.,Department of Social & Behavioral Sciences, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA.,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Golareh Agha
- Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Charles B Eaton
- Department of Epidemiology, Brown School of Public Health, Providence, RI, 02912, USA.,Department of Family Medicine, Brown University Warren Alpert Medical School, Providence, RI, 02912, USA
| | - Stephen L Buka
- Department of Epidemiology, Brown School of Public Health, Providence, RI, 02912, USA
| | - Yen-Tsung Huang
- Department of Epidemiology, Brown School of Public Health, Providence, RI, 02912, USA.,Department of Biostatistics, Brown School of Public Health, Providence, RI, 02912, USA.,Institute of Statistical Science, Academia Sinica, Taipei, 11529, Taiwan
| |
Collapse
|
45
|
Zhang L, Ning Y, Li P, Guo H, Zan L. Tissue Expression Analysis and Characterization of Smad3 Promoter in Bovine Myoblasts and Preadipocytes. DNA Cell Biol 2018; 37:551-559. [PMID: 29672161 PMCID: PMC5985903 DOI: 10.1089/dna.2018.4152] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 03/07/2018] [Accepted: 03/20/2018] [Indexed: 11/12/2022] Open
Abstract
The transforming growth factor-β (TGFβ) pathway plays many key roles in regulating numerous biological processes. In addition, the effects of TGFβ are mediated by the transcription factor Smad3. However, the regulation of Smad3 activity is not well understood. In the present study, quantitative real-time PCR revealed that the Smad3 gene was expressed ubiquitously in 11 bovine tissues and displayed different expression patterns between muscle and adipose tissue. We further explored the expression and regulation of Smad3 gene by cloning the bovine Smad3 gene promoter; a dual-luciferase reporter assay identified that the core promoter region -337 to -41 bp was located in a CpG island. In addition, mutational analyses and electrophoretic mobility shift assays provided evidence that the KLF6, KLF15, MZF1, and KLF7 binding sites within the Smad3 promoter were responsible for the regulation of Smad3 transcription. These findings were confirmed by executing further RNA interference assays in bovine myoblasts and preadipocytes, which indicated that KLF6, KLF15, MZF1, and KLF7 are important transcriptional activators of Smad3 in both adipose and muscle tissue. These results will provide an important basis for an improved understanding of the TGFβ pathway and new insights in cattle breeding.
Collapse
Affiliation(s)
- Le Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yue Ning
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Peiwei Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hongfang Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
- National Beef Cattle Improvement Center, Yangling, China
| |
Collapse
|
46
|
Sharma N, Akkoyunlu M, Rabin RL. Macrophages-common culprit in obesity and asthma. Allergy 2018; 73:1196-1205. [PMID: 29178573 DOI: 10.1111/all.13369] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2017] [Indexed: 12/29/2022]
Abstract
Macrophages are essential innate immune cells that also regulate local metabolism. Endogenous or exogenous stimuli may polarize macrophages toward phenotypes that serve distinct innate immunological metabolic functions. IFN-γ or lipopolysaccharide (LPS) polarizes macrophages toward the M1, or "classically activated" phenotype that participates in defense against intracellular pathogens. IL-4, IL-13, or chitin polarizes macrophages toward the M2, or "alternatively activated" phenotype, which defends against multicellular nematodes and fungi. As macrophages polarize in local environments, M1 and M2 macrophages may coexist in different organs and may differentially affect asthma and obesity, two comorbid diseases where polarized macrophages contribute to their pathogenesis. While M1 macrophages are considered beneficial in asthma and contribute to the pathology of obesity, M2 macrophages contribute to the pathology of asthma, but limit metabolic syndrome associated with obesity. Here, we discuss the roles for M1 and M2 macrophages in asthma and obesity, and propose a model by which M1-mediated inflammation in adipose tissue enhances M2-mediated inflammation in the asthmatic lung.
Collapse
Affiliation(s)
- N. Sharma
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| | - M. Akkoyunlu
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| | - R. L. Rabin
- Division of Bacterial, Parasitic and Allergenic Products Center for Biologics Evaluation and Research Office of Vaccines Research and Review U.S. Food and Drug Administration Silver Spring MD USA
| |
Collapse
|
47
|
Chemical compound-based direct reprogramming for future clinical applications. Biosci Rep 2018; 38:BSR20171650. [PMID: 29739872 PMCID: PMC5938430 DOI: 10.1042/bsr20171650] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/29/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022] Open
Abstract
Recent studies have revealed that a combination of chemical compounds enables direct reprogramming from one somatic cell type into another without the use of transgenes by regulating cellular signaling pathways and epigenetic modifications. The generation of induced pluripotent stem (iPS) cells generally requires virus vector-mediated expression of multiple transcription factors, which might disrupt genomic integrity and proper cell functions. The direct reprogramming is a promising alternative to rapidly prepare different cell types by bypassing the pluripotent state. Because the strategy also depends on forced expression of exogenous lineage-specific transcription factors, the direct reprogramming in a chemical compound-based manner is an ideal approach to further reduce the risk for tumorigenesis. So far, a number of reported research efforts have revealed that combinations of chemical compounds and cell-type specific medium transdifferentiate somatic cells into desired cell types including neuronal cells, glial cells, neural stem cells, brown adipocytes, cardiomyocytes, somatic progenitor cells, and pluripotent stem cells. These desired cells rapidly converted from patient-derived autologous fibroblasts can be applied for their own transplantation therapy to avoid immune rejection. However, complete chemical compound-induced conversions remain challenging particularly in adult human-derived fibroblasts compared with mouse embryonic fibroblasts (MEFs). This review summarizes up-to-date progress in each specific cell type and discusses prospects for future clinical application toward cell transplantation therapy.
Collapse
|
48
|
Smad proteins differentially regulate obesity-induced glucose and lipid abnormalities and inflammation via class-specific control of AMPK-related kinase MPK38/MELK activity. Cell Death Dis 2018; 9:471. [PMID: 29700281 PMCID: PMC5920110 DOI: 10.1038/s41419-018-0489-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 12/16/2022]
Abstract
Smad proteins have been implicated in metabolic processes, but little is known about how they regulate metabolism. Because Smad 2, 3, 4, and 7 have previously been shown to interact with murine protein serine-threonine kinase 38 (MPK38), an AMP-activated protein kinase (AMPK)-related kinase that has been implicated in obesity-associated metabolic defects, we investigated whether Smad proteins regulate metabolic processes via MPK38. Smads2/3/4 increased, but Smad7 decreased, MPK38-mediated apoptosis signal-regulating kinase-1 (ASK1)/transforming growth factor-β (TGF-β)/p53 signaling. However, MPK38-mediated phosphorylation-defective Smad mutants (Smad2 S245A, Smad3 S204A, Smad4 S343A, and Smad7 T96A) had no such effect. In addition, Smads2/3/4 increased, but Smad7 decreased, the stability of MPK38. Consistent with this, Smads2/3/4 attenuated complex formation between MPK38 and its negative regulator thioredoxin (Trx), whereas Smad7 increased this complex formation. However, an opposite effect was observed on complex formation between MPK38 and its positive regulator zinc-finger-like protein 9 (ZPR9). When Smads were overexpressed in high-fat diet (HFD)-fed obese mice using an adenoviral delivery system, Smads2/3/4 improved, but Smad7 worsened, obesity-associated metabolic parameters and inflammation in a MPK38 phosphorylation-dependent manner. These findings suggest that Smad proteins have class-specific impacts on obesity-associated metabolism by differentially regulating MPK38 activity in diet-induced obese mice.
Collapse
|
49
|
Rotini A, Martínez-Sarrà E, Duelen R, Costamagna D, Di Filippo ES, Giacomazzi G, Grosemans H, Fulle S, Sampaolesi M. Aging affects the in vivo regenerative potential of human mesoangioblasts. Aging Cell 2018; 17. [PMID: 29397577 PMCID: PMC5847873 DOI: 10.1111/acel.12714] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2017] [Indexed: 01/29/2023] Open
Abstract
Sarcopenia is the age‐related loss of muscle mass, strength, and function. Although the role of human satellite cells (SCs) as adult skeletal muscle stem cells has been deeply investigated, little is known about the impact of aging on muscle interstitial stem cells. Here, we isolated the non‐SC CD56– fraction from human muscle biopsies of young and elderly subjects. The elderly interstitial cell population contained a higher number of CD15+ and PDGFRα+ cells when compared to young samples. In addition, we found that the CD56–/ALP+ cells were well represented as a multipotent stem cell population inside the CD56– fraction. CD56–/ALP+/CD15– cells were clonogenic, and since they were myogenic and expressed NG2, α‐SMA and PDGFRβ can be considered mesoangioblasts (MABs). Interestingly, elderly MABs displayed a dramatic impairment in the myogenic differentiation ability in vitro and when transplanted in dystrophic immunodeficient Sgcb‐null Rag2‐null γc‐null mice. In addition, elderly MABs proliferated less, but yet retained other multilineage capabilities. Overall, our results indicate that aging negatively impacted on the regenerative potential of MABs and this should be carefully considered for potential therapeutic applications of MABs.
Collapse
Affiliation(s)
- Alessio Rotini
- Translational Cardiomyology Laboratory; Stem Cell Institute of Leuven; Unit of Stem Cell Research; Cluster of Stem Cell and Developmental Biology; Department of Development and Regeneration; University of Leuven; Leuven Belgium
- Department of Neuroscience, Imaging and Clinical Sciences; University “G. d'Annunzio” Chieti-Pescara; Chieti Italy
- Interuniversity Institute of Myology; Chieti Italy
| | - Ester Martínez-Sarrà
- Translational Cardiomyology Laboratory; Stem Cell Institute of Leuven; Unit of Stem Cell Research; Cluster of Stem Cell and Developmental Biology; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Robin Duelen
- Translational Cardiomyology Laboratory; Stem Cell Institute of Leuven; Unit of Stem Cell Research; Cluster of Stem Cell and Developmental Biology; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Domiziana Costamagna
- Translational Cardiomyology Laboratory; Stem Cell Institute of Leuven; Unit of Stem Cell Research; Cluster of Stem Cell and Developmental Biology; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Ester Sara Di Filippo
- Department of Neuroscience, Imaging and Clinical Sciences; University “G. d'Annunzio” Chieti-Pescara; Chieti Italy
- Interuniversity Institute of Myology; Chieti Italy
| | - Giorgia Giacomazzi
- Translational Cardiomyology Laboratory; Stem Cell Institute of Leuven; Unit of Stem Cell Research; Cluster of Stem Cell and Developmental Biology; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Hanne Grosemans
- Translational Cardiomyology Laboratory; Stem Cell Institute of Leuven; Unit of Stem Cell Research; Cluster of Stem Cell and Developmental Biology; Department of Development and Regeneration; University of Leuven; Leuven Belgium
| | - Stefania Fulle
- Department of Neuroscience, Imaging and Clinical Sciences; University “G. d'Annunzio” Chieti-Pescara; Chieti Italy
- Interuniversity Institute of Myology; Chieti Italy
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory; Stem Cell Institute of Leuven; Unit of Stem Cell Research; Cluster of Stem Cell and Developmental Biology; Department of Development and Regeneration; University of Leuven; Leuven Belgium
- Interuniversity Institute of Myology; Chieti Italy
- Human Anatomy Unit; Department of Public Health, Experimental and Forensic Medicine; University of Pavia; Pavia Italy
| |
Collapse
|
50
|
Yoo KH, Yim HE, Bae ES, Hong YS. Genetic Contributions to Childhood Obesity: Association of Candidate Gene Polymorphisms and Overweight/Obesity in Korean Preschool Children. J Korean Med Sci 2017; 32:1997-2004. [PMID: 29115082 PMCID: PMC5680499 DOI: 10.3346/jkms.2017.32.12.1997] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/03/2017] [Indexed: 01/26/2023] Open
Abstract
This study was aimed to investigate the association of candidate gene polymorphisms and obesity or overweight in young Korean children. A total of 190 Korean preschool children (96 control, 48 overweight, and 46 obese children) were genotyped for the angiotensin converting enzyme (ACE) insertion (I)/deletion (D), angiotensin II type 2 receptor (AT2) C3123A, transforming growth factor (TGF)-β1 T869C, vascular endothelial growth factor (VEGF) T460C, and tumor necrosis factor (TNF)-α G308A polymorphisms. No differences were found among the groups with respect to age, sex, birth weight, blood pressure levels, and serum concentrations of glucose and total cholesterol. Obese children showed a higher incidence of ACE DD genotype and D allelic frequency compared to the controls (odds ratio [OR], 2.7, 95% confidence interval [CI], 1.01-7.21; OR, 2.5, 95% CI, 1.49-4.19; all P < 0.05). The frequency of TC genotype and C allele in the TGF-β1 T869C polymorphism (OR, 2.08, 95% CI, 1.01-4.27; OR, 1.93, 95% CI, 1.15-3.21) and that in the VEGF T460C polymorphism (OR, 2.5, 95% CI, 1.19-5.28; OR, 2.15, 95% CI, 1.26-3.68) was also higher in obese children than in control subjects (all P < 0.05). Overweight children exhibited a higher frequency of the A allele in the AT2 C3123A polymorphism compared to the controls (OR, 1.72, 95% CI, 1.03-2.88, P < 0.05). There were no differences in the TNF-α G308A polymorphism among the groups. The ACE I/D, AT2 C3123A, TGF-β1 T869C, and VEGF T460C polymorphisms can affect susceptibility to obesity or overweight in Korean children.
Collapse
Affiliation(s)
- Kee Hwan Yoo
- Department of Pediatrics, Korea University Ansan Hospital, Ansan, Korea
| | - Hyung Eun Yim
- Department of Pediatrics, Korea University Ansan Hospital, Ansan, Korea.
| | - Eun Soo Bae
- Department of Pediatrics, Korea University Ansan Hospital, Ansan, Korea
| | - Young Sook Hong
- Department of Pediatrics, Korea University Ansan Hospital, Ansan, Korea
| |
Collapse
|