1
|
Liu X, Liu K, Wang Y, Meng X, Wang Q, Tao S, Xu Q, Shen X, Gao X, Hong S, Jin H, Wang JQ, Wang D, Lu L, Meng Z, Wang L. SWI/SNF chromatin remodeling factor BAF60b restrains inflammatory diseases by affecting regulatory T cell migration. Cell Rep 2024; 43:114458. [PMID: 38996070 DOI: 10.1016/j.celrep.2024.114458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/21/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Regulatory T (Treg) cells play a critical regulatory role in the immune system by suppressing excessive immune responses and maintaining immune balance. The effective migration of Treg cells is crucial for controlling the development and progression of inflammatory diseases. However, the mechanisms responsible for directing Treg cells into the inflammatory tissue remain incompletely elucidated. In this study, we identified BAF60b, a subunit of switch/sucrose nonfermentable (SWI/SNF) chromatin remodeling complexes, as a positive regulator of Treg cell migration that inhibits the progression of inflammation in experimental autoimmune encephalomyelitis (EAE) and colitis animal models. Mechanistically, transcriptome and genome-wide chromatin-landscaped analyses demonstrated that BAF60b interacts with the transcription factor RUNX1 to promote the expression of CCR9 on Treg cells, which in turn affects their ability to migrate to inflammatory tissues. Our work provides insights into the essential role of BAF60b in regulating Treg cell migration and its impact on inflammatory diseases.
Collapse
MESH Headings
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Animals
- Cell Movement
- Mice
- Mice, Inbred C57BL
- Inflammation/pathology
- Inflammation/metabolism
- Chromatin Assembly and Disassembly
- Chromosomal Proteins, Non-Histone/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Humans
- Transcription Factors/metabolism
- Core Binding Factor Alpha 2 Subunit/metabolism
- Core Binding Factor Alpha 2 Subunit/genetics
- Colitis/metabolism
- Colitis/pathology
- Colitis/immunology
- Colitis/genetics
Collapse
Affiliation(s)
- Xiaoqian Liu
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China; Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kuai Liu
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China; Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuxi Wang
- Laboratory Animal Center, Zhejiang University, Hangzhou 310058, China
| | - Xiaoyu Meng
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China; Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qianqian Wang
- Laboratory Animal Center, Zhejiang University, Hangzhou 310058, China
| | - Sijue Tao
- Laboratory Animal Center, Zhejiang University, Hangzhou 310058, China
| | - Qianying Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xin Shen
- Co-Facility Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xianzhi Gao
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311100, China; Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shenghui Hong
- Laboratory Animal Center, Zhejiang University, Hangzhou 310058, China
| | - Huihui Jin
- Laboratory Animal Center, Zhejiang University, Hangzhou 310058, China
| | - James Q Wang
- Zhejiang University School of Medicine, Hangzhou 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China
| | - Di Wang
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Linrong Lu
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhuoxian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology, School of Medicine, Second Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
| | - Lie Wang
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311100, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311100, China; Zhejiang University School of Medicine, Hangzhou 310058, China; Laboratory Animal Center, Zhejiang University, Hangzhou 310058, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China; Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
2
|
Basurto-Cayuela L, Guerrero-Martínez JA, Gómez-Marín E, Sánchez-Escabias E, Escaño-Maestre M, Ceballos-Chávez M, Reyes JC. SWI/SNF-dependent genes are defined by their chromatin landscape. Cell Rep 2024; 43:113855. [PMID: 38427563 DOI: 10.1016/j.celrep.2024.113855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 11/23/2023] [Accepted: 02/08/2024] [Indexed: 03/03/2024] Open
Abstract
SWI/SNF complexes are evolutionarily conserved, ATP-dependent chromatin remodeling machines. Here, we characterize the features of SWI/SNF-dependent genes using BRM014, an inhibitor of the ATPase activity of the complexes. We find that SWI/SNF activity is required to maintain chromatin accessibility and nucleosome occupancy for most enhancers but not for most promoters. SWI/SNF activity is needed for expression of genes with low to medium levels of expression that have promoters with (1) low chromatin accessibility, (2) low levels of active histone marks, (3) high H3K4me1/H3K4me3 ratio, (4) low nucleosomal phasing, and (5) enrichment in TATA-box motifs. These promoters are mostly occupied by the canonical Brahma-related gene 1/Brahma-associated factor (BAF) complex. These genes are surrounded by SWI/SNF-dependent enhancers and mainly encode signal transduction, developmental, and cell identity genes (with almost no housekeeping genes). Machine-learning models trained with different chromatin characteristics of promoters and their surrounding regulatory regions indicate that the chromatin landscape is a determinant for establishing SWI/SNF dependency.
Collapse
Affiliation(s)
- Laura Basurto-Cayuela
- Genome Biology Department, Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO), Av. Americo Vespucio, 41092 Seville, Spain
| | - José A Guerrero-Martínez
- Genome Biology Department, Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO), Av. Americo Vespucio, 41092 Seville, Spain
| | - Elena Gómez-Marín
- Genome Biology Department, Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO), Av. Americo Vespucio, 41092 Seville, Spain
| | - Elena Sánchez-Escabias
- Genome Biology Department, Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO), Av. Americo Vespucio, 41092 Seville, Spain
| | - María Escaño-Maestre
- Genome Biology Department, Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO), Av. Americo Vespucio, 41092 Seville, Spain
| | - María Ceballos-Chávez
- Genome Biology Department, Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO), Av. Americo Vespucio, 41092 Seville, Spain
| | - José C Reyes
- Genome Biology Department, Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO), Av. Americo Vespucio, 41092 Seville, Spain.
| |
Collapse
|
3
|
Chen Z, Zhang P, Liu T, Qiu X, Li S, Lin JD. Neuregulin 4 mediates the metabolic benefits of mild cold exposure by promoting beige fat thermogenesis. JCI Insight 2024; 9:e172957. [PMID: 38015639 PMCID: PMC10906454 DOI: 10.1172/jci.insight.172957] [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: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Interorgan crosstalk via secreted hormones and metabolites is a fundamental aspect of mammalian metabolic physiology. Beyond the highly specialized endocrine cells, peripheral tissues are emerging as an important source of metabolic hormones that influence energy and nutrient metabolism and contribute to disease pathogenesis. Neuregulin 4 (Nrg4) is a fat-derived hormone that protects mice from nonalcoholic steatohepatitis (NASH) and NASH-associated liver cancer by shaping hepatic lipid metabolism and the liver immune microenvironment. Despite its enriched expression in brown fat, whether NRG4 plays a role in thermogenic response and mediates the metabolic benefits of cold exposure are areas that remain unexplored. Here we show that Nrg4 expression in inguinal white adipose tissue (iWAT) is highly responsive to chronic cold exposure. Nrg4 deficiency impairs beige fat induction and renders mice more susceptible to diet-induced metabolic disorders under mild cold conditions. Using mice with adipocyte and hepatocyte-specific Nrg4 deletion, we reveal that adipose tissue-derived NRG4, but not hepatic NRG4, is essential for beige fat induction following cold acclimation. Furthermore, treatment with recombinant NRG4-Fc fusion protein promotes beige fat induction in iWAT and improves metabolic health in mice with diet-induced obesity. These findings highlight a critical role of NRG4 in mediating beige fat induction and preserving metabolic health under mild cold conditions.
Collapse
|
4
|
Ludzki AC, Hansen M, Zareifi D, Jalkanen J, Huang Z, Omar-Hmeadi M, Renzi G, Klingelhuber F, Boland S, Ambaw YA, Wang N, Damdimopoulos A, Liu J, Jernberg T, Petrus P, Arner P, Krahmer N, Fan R, Treuter E, Gao H, Rydén M, Mejhert N. Transcriptional determinants of lipid mobilization in human adipocytes. SCIENCE ADVANCES 2024; 10:eadi2689. [PMID: 38170777 PMCID: PMC10776019 DOI: 10.1126/sciadv.adi2689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
Defects in adipocyte lipolysis drive multiple aspects of cardiometabolic disease, but the transcriptional framework controlling this process has not been established. To address this, we performed a targeted perturbation screen in primary human adipocytes. Our analyses identified 37 transcriptional regulators of lipid mobilization, which we classified as (i) transcription factors, (ii) histone chaperones, and (iii) mRNA processing proteins. On the basis of its strong relationship with multiple readouts of lipolysis in patient samples, we performed mechanistic studies on one hit, ZNF189, which encodes the zinc finger protein 189. Using mass spectrometry and chromatin profiling techniques, we show that ZNF189 interacts with the tripartite motif family member TRIM28 and represses the transcription of an adipocyte-specific isoform of phosphodiesterase 1B (PDE1B2). The regulation of lipid mobilization by ZNF189 requires PDE1B2, and the overexpression of PDE1B2 is sufficient to attenuate hormone-stimulated lipolysis. Thus, our work identifies the ZNF189-PDE1B2 axis as a determinant of human adipocyte lipolysis and highlights a link between chromatin architecture and lipid mobilization.
Collapse
Affiliation(s)
- Alison C. Ludzki
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Mattias Hansen
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Danae Zareifi
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Jutta Jalkanen
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Zhiqiang Huang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83 Stockholm, Sweden
| | - Muhmmad Omar-Hmeadi
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Gianluca Renzi
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Felix Klingelhuber
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sebastian Boland
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Yohannes A. Ambaw
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Cell Biology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Na Wang
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Anastasios Damdimopoulos
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83 Stockholm, Sweden
| | - Jianping Liu
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Tomas Jernberg
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, SE-182 88 Stockholm, Sweden
| | - Paul Petrus
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Peter Arner
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany
- Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83 Stockholm, Sweden
| | - Eckardt Treuter
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83 Stockholm, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, SE-141 83 Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| | - Niklas Mejhert
- Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden
| |
Collapse
|
5
|
Zhao Y, Liu Y, Zhao G, Lu H, Liu Y, Xue C, Chang Z, Liu H, Deng Y, Liang W, Wang H, Rom O, Garcia-Barrio MT, Zhu T, Guo Y, Chang L, Lin J, Chen YE, Zhang J. Myeloid BAF60a deficiency alters metabolic homeostasis and exacerbates atherosclerosis. Cell Rep 2023; 42:113171. [PMID: 37768825 PMCID: PMC10842557 DOI: 10.1016/j.celrep.2023.113171] [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: 03/20/2023] [Revised: 08/15/2023] [Accepted: 09/07/2023] [Indexed: 09/30/2023] Open
Abstract
Atherosclerosis, a leading health concern, stems from the dynamic involvement of immune cells in vascular plaques. Despite its significance, the interplay between chromatin remodeling and transcriptional regulation in plaque macrophages is understudied. We discovered the reduced expression of Baf60a, a component of the switch/sucrose non-fermentable (SWI/SNF) chromatin remodeling complex, in macrophages from advanced plaques. Myeloid-specific Baf60a deletion compromised mitochondrial integrity and heightened adhesion, apoptosis, and plaque development. BAF60a preserves mitochondrial energy homeostasis under pro-atherogenic stimuli by retaining nuclear respiratory factor 1 (NRF1) accessibility at critical genes. Overexpression of BAF60a rescued mitochondrial dysfunction in an NRF1-dependent manner. This study illuminates the BAF60a-NRF1 axis as a mitochondrial function modulator in atherosclerosis, proposing the rejuvenation of perturbed chromatin remodeling machinery as a potential therapeutic target.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yuhao Liu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Guizhen Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Haocheng Lu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Pharmacology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yaozhong Liu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Chao Xue
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Ziyi Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Hongyu Liu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yongjie Deng
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Wenying Liang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Huilun Wang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Oren Rom
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Pathology and Translational Pathobiology, Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA 71103, USA
| | - Minerva T Garcia-Barrio
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Tianqing Zhu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yanhong Guo
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Lin Chang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jiandie Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
6
|
Zhang L, Ma P, Wang Z, Xu T, Lam SM, Shui G, Wang Y, Xie J, Qiang G. Multiomics Approaches Identify Biomarkers for BAT Thermogenesis. J Proteome Res 2023; 22:3332-3347. [PMID: 37616386 DOI: 10.1021/acs.jproteome.3c00423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Brown adipose tissue (BAT) thermogenesis confers beneficial effects on metabolic diseases such as obesity and type-2 diabetes. Nevertheless, the mechanism and lipid driving the process that evokes this response have not been investigated yet. Here, a multiomics approach of integrative transcriptomics and lipidomics is used to explore the mechanism of regulating thermogenesis in BAT and providing promising lipid biomarkers and biomarker genes for thermogenic activators as antiobesity drugs. Lipidomics analysis demonstrated that a high abundance of glycerophospholipids and sphingolipids was more significant in BAT than in WAT. Enrichment analysis of upregulated DEGs between WAT and BAT screened suggested that the differences were mainly involved in lipid metabolism. Besides, β3-adrenergic agonist stimulation reduced the levels of TAG and DAG and increased the content of PC, PE, CL, and LPC and expression of genes involved in thermogenesis, fatty acid elongation, and glycerophospholipid metabolism in BAT. In this study, based on interpreting the inherent characterization of BAT as thermogenic tissue through comparison with WAT as fat storage tissue, adrenergic stimulation-induced BAT thermogenesis further identified specific lipid biomarkers (7 TAG species, 10 PC species, 1 LPC species, and 1 CL species) and Elovl3 and Crat gene biomarkers, which may provide targets for combating obesity by boosting BAT thermogenesis.
Collapse
Affiliation(s)
- Li Zhang
- Inner Mongolia Clinical College, Inner Mongolia Medical University, Hohhot 010110, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Peng Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Zijing Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Tianshu Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuzhen Wang
- College of Life Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Jiming Xie
- Inner Mongolia Clinical College, Inner Mongolia Medical University, Hohhot 010110, China
- Clinical Laboratory, Inner Mongolia People's Hospital, Hohhot 010020, China
| | - Guifen Qiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| |
Collapse
|
7
|
Liu X, Huang C, Jiang T, Sun X, Zhan S, Zhong T, Guo J, Dai D, Wang Y, Li L, Zhang H, Wang L. LncDGAT2 is a novel positive regulator of the goat adipocyte thermogenic gene program. Int J Biol Macromol 2023; 245:125465. [PMID: 37355065 DOI: 10.1016/j.ijbiomac.2023.125465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/29/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023]
Abstract
Brown and beige adipose thermogenesis are important for newborn mammals to maintain their body temperature. In addition, these thermogenic fats are regulated by multiple molecular interactions. How the long non-coding RNAs (lncRNAs) regulate adipose thermogenesis in newborn mammals upon cold exposure remains unexplored. Here, we identified lncRNAs induced by cold exposure in brown adipose tissue (BAT) of newborn goats and found that lncDGAT2 was enriched in BAT after cold exposure. Functional studies revealed that lncDGAT2 promoted brown and white adipocyte differentiation as well as thermogenic gene expression. Additionally, PRDM4 directly bound the lncDGAT2 promoter to activate the transcription of lncDGAT2 and the PRDM4-lncDGAT2 axis was essential for the brown adipocyte thermogenic gene program. These findings provide evidence for lncRNA and transcription factor regulatory functions in controlling adipose thermogenesis and energy metabolism of newborn goats.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Chunhua Huang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Tingting Jiang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Xueliang Sun
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Siyuan Zhan
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Tao Zhong
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Dinghui Dai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Li Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Linjie Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| |
Collapse
|
8
|
Shen H, He T, Wang S, Hou L, Wei Y, Liu Y, Mo C, Zhao Z, You W, Guo H, Li B. SOX4 promotes beige adipocyte-mediated adaptive thermogenesis by facilitating PRDM16-PPARγ complex. Theranostics 2022; 12:7699-7716. [PMID: 36451857 PMCID: PMC9706582 DOI: 10.7150/thno.77102] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
Brown and beige fat protect against cold environments and obesity by catabolizing stored energy to generate heat. This process is achieved by controlling thermogenesis-related gene expression and the development of brown/beige fat through the induction of transcription factors, most notably PPARγ. However, the cofactors that induce the expression of thermogenic genes with PPARγ are still not well understood. In this study, we explored the role of SOX4 in adaptive thermogenesis and its relationship with PPARγ. Methods: Whole transcriptome deep sequencing (RNA-seq) analysis of inguinal subcutaneous white adipose tissue (iWAT) after cold stimulation was performed to identify genes with differential expression in mice. Indirect calorimetry detected oxygen consumption rate and heat generation. mRNA levels were analyzed by qPCR assays. Proteins were detected by immunoblotting and immunofluorescence. Interaction of proteins was detected by endogenous and exogenous Co-IP. ChIP-qPCR, FAIRE assay and luciferase reporter assays were used to investigate transcriptional regulation. Results: SOX4 was identified as the main transcriptional effector of thermogenesis. Mice with either adipocyte-specific or UCP1+ cells deletion of SOX4 exhibited significant cold intolerance, decreased energy expenditure, and beige adipocyte formation, which was attributed to decreased thermogenic gene expression. In addition, these mice developed obesity on a high-fat diet, with severe hepatic steatosis, insulin resistance, and inflammation. At the cell level, loss of SOX4 from preadipocytes inhibited the development of beige adipocytes, and loss of SOX4 from mature beige adipocytes reduced the expression of thermogenesis-related genes and energy metabolism. Mechanistically, SOX4 stimulated the transcriptional activity of Ucp1 by binding to PPARγ and activating its transcriptional function. These actions of SOX4 were, at least partly, mediated by recruiting PRDM16 to PPARγ, thus forming a transcriptional complex to elevate the expression of thermogenic genes. Conclusion: SOX4, as a coactivator of PPARγ, drives the thermogenic gene expression program and thermogenesis of beige fat, promoting energy expenditure. It has important physiological significance in resisting cold and obesity.
Collapse
Affiliation(s)
- Huanming Shen
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Ting He
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Shuai Wang
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Lingfeng Hou
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Yixin Wei
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Yunjia Liu
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Chunli Mo
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Zehang Zhao
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - WeiXin You
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China
| | - Huiling Guo
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China.,✉ Corresponding authors: Dr. Huiling Guo School of Life Sciences, Xiamen University, Xiang'an District, Xiamen, Fujian, China, 361102; Tel: 86-592-2186717; E-mail: . Dr. Boan Li School of Life Sciences, Xiamen University, Xiang'an District, Xiamen, Fujian, China, 361102; Tel: 86-592-2186717; E-mail:
| | - Boan Li
- State key laboratory of cellular stress biology, innovation center for cell signaling network and engineering research center of molecular diagnostics of the ministry of education, school of life sciences, Xiamen university, Xiamen 361100, Fujian, China.,Lead Contact.,✉ Corresponding authors: Dr. Huiling Guo School of Life Sciences, Xiamen University, Xiang'an District, Xiamen, Fujian, China, 361102; Tel: 86-592-2186717; E-mail: . Dr. Boan Li School of Life Sciences, Xiamen University, Xiang'an District, Xiamen, Fujian, China, 361102; Tel: 86-592-2186717; E-mail:
| |
Collapse
|
9
|
Dalamaga M, Liu J. A chromatin remodeling checkpoint of diet-induced macrophage activation in adipose tissue. Metabol Open 2022; 15:100204. [PMID: 35990770 PMCID: PMC9386063 DOI: 10.1016/j.metop.2022.100204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 12/25/2022] Open
Abstract
The interplay between the environment and the immune cells is linked to metabolic homeostasis under physiologic and pathophysiologic conditions. Diabetes mellitus type 2 (T2D) is considered an immune-related inflammatory disorder, in which the adipose tissue macrophages (ATMs) are key players orchestrating metabolic chronic meta-inflammation and contributing to the pathogenesis of metabolic disease. However, the molecular regulators that integrate the environmental signals to control ATM activation and adipose inflammation during obesity and T2D remain unclear. Epigenetic mechanisms constitute important parameters in metabolic homeostasis, obesity and T2D via the integration of the environmental factors to the transcriptional regulation of gene programs. In a very recent study published in Diabetes by Kong et al., BAF60a has been identified as a key chromatin remodeling checkpoint factor that associates obesity-associated stress signals with meta-inflammation and systemic homeostasis. Furthermore, this work uncovers Atf3 as an important downstream effector in BAF60a-mediated chromatin remodeling and transcriptional reprogramming of macrophage activation in adipose tissue. The findings of this research may contribute to the development of new therapeutic approaches for obesity-induced metabolic inflammation and associated metabolic disorders.
Collapse
Affiliation(s)
- Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, Goudi, 11527, Athens, Greece
| | - Junli Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University Affiliated 6th People's Hospital, Shanghai Diabetes Institute, Shanghai, China
| |
Collapse
|
10
|
Integrated Analysis of Transcriptome, microRNAs, and Chromatin Accessibility Revealed Potential Early B-Cell Factor1-Regulated Transcriptional Networks during the Early Development of Fetal Brown Adipose Tissues in Rabbits. Cells 2022; 11:cells11172675. [PMID: 36078081 PMCID: PMC9454897 DOI: 10.3390/cells11172675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
In domestic mammals, cold stress decreases the survival rate of newborns and increases the cost of management. Brown adipose tissue (BAT) is the main thermogenic organ for cubs, and well-developed fetal BAT (FBAT) is beneficial for newborns to maintain core temperatures during the first several days of life. However, our knowledge of the epigenetic mechanisms during the early development of FBAT remains largely unknown. Rabbits (Oryctolagus cuniculus) are economically important domestic animals. In this study, a histological analysis shows that the tissue content, thermogenic capacity, and lipid content of FBAT dramatically increases from gestational day 21 (G21) to gestational day 24 (G24) in rabbits. RNA-seq, microRNA-seq (miRNA-seq), and the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) show that many genes, miRNAs, and chromatin-accessible regions (referred to as peaks) were identified as significantly changed from G21 to G24, respectively. The upregulated genes from G21 to G24 were significantly enriched in the mitochondrial metabolism and thermogenesis-related signal pathways. The integrated analysis of transcriptome and chromatin accessibility reveals that the peaks in the promoters have a more regulatory effect than peaks in other genomic elements on the expression of their nearby genes in FBATs. The upregulated genes that are associated with increased chromatin accessibility in the promoter regions are involved in the energy metabolism-related signaling pathways. The genes that have a greater tendency to be regulated by miRNAs than the chromatin accessibility in gene promoters are involved in the apelin, insulin, and endocytosis signaling pathways. Furthermore, genome-wide transcription factor (TF) footprinting analysis identifies early B-cell factor1 (EBF1) as playing a key role during early FBAT development. The carbon metabolism, citrate cycle, and PPAR signaling pathways are significantly enriched by the predicted EBF1-regulated cascade TF-network. In conclusion, our work provides a framework for understanding epigenetics regulatory mechanisms underlying the early development of FBAT and identifies potential TF involved in the early development of FBAT in rabbits.
Collapse
|
11
|
Wang RR, Qiu X, Pan R, Fu H, Zhang Z, Wang Q, Chen H, Wu QQ, Pan X, Zhou Y, Shan P, Wang S, Guo G, Zheng M, Zhu L, Meng ZX. Dietary intervention preserves β cell function in mice through CTCF-mediated transcriptional reprogramming. J Exp Med 2022; 219:213256. [PMID: 35652891 DOI: 10.1084/jem.20211779] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 04/04/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β cell plasticity is the primary determinant of disease progression and remission of type 2 diabetes (T2D). However, the dynamic nature of β cell adaptation remains elusive. Here, we establish a mouse model exhibiting the compensation-to-decompensation adaptation of β cell function in response to increasing duration of high-fat diet (HFD) feeding. Comprehensive islet functional and transcriptome analyses reveal a dynamic orchestration of transcriptional networks featuring temporal alteration of chromatin remodeling. Interestingly, prediabetic dietary intervention completely rescues β cell dysfunction, accompanied by a remarkable reversal of HFD-induced reprogramming of islet chromatin accessibility and transcriptome. Mechanistically, ATAC-based motif analysis identifies CTCF as the top candidate driving dietary intervention-induced preservation of β cell function. CTCF expression is markedly decreased in β cells from obese and diabetic mice and humans. Both dietary intervention and AAV-mediated restoration of CTCF expression ameliorate β cell dysfunction ex vivo and in vivo, through transducing the lipid toxicity and inflammatory signals to transcriptional reprogramming of genes critical for β cell glucose metabolism and stress response.
Collapse
Affiliation(s)
- Ruo-Ran Wang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinyuan Qiu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Ran Pan
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongxing Fu
- Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ziyin Zhang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qintao Wang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haide Chen
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qing-Qian Wu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaowen Pan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanping Zhou
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pengfei Shan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Tianjin, China.,NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Chronic Disease Research Institute, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
A distinct role of STING in regulating glucose homeostasis through insulin sensitivity and insulin secretion. Proc Natl Acad Sci U S A 2022; 119:2101848119. [PMID: 35145023 PMCID: PMC8851542 DOI: 10.1073/pnas.2101848119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
The role of STING in maintaining glucose homeostasis remains unknown. Herein, using global and β-cell–specific STING knockout mouse models, we revealed a distinct role of STING in the regulation of glucose homeostasis through β-cells and peripheral tissues. Specially, while global STING knockout beneficially alleviated insulin resistance and glucose intolerance induced by high-fat diet, it surprisingly impaired islet glucose-stimulated insulin secretion (GSIS). Further analyses revealed that STING deficiency down-regulated expression of β-cell key transcription factor Pax6, impairing Pax6 nuclear localization and binding activity to the promoters of its target genes, including Glut2 and Abcc8, causing impaired GSIS. These data highlight pathophysiological significance of fine-tuned STING signaling in β-cells and insulin target tissues for maintaining glucose homeostasis. Insulin resistance and β-cell dysfunction are two main molecular bases yet to be further elucidated for type 2 diabetes (T2D). Accumulating evidence indicates that stimulator of interferon genes (STING) plays an important role in regulating insulin sensitivity. However, its function in β-cells remains unknown. Herein, using global STING knockout (STING−/−) and β-cell–specific STING knockout (STING-βKO) mouse models, we revealed a distinct role of STING in the regulation of glucose homeostasis through peripheral tissues and β-cells. Specially, although STING−/− beneficially alleviated insulin resistance and glucose intolerance induced by high-fat diet, it surprisingly impaired islet glucose-stimulated insulin secretion (GSIS). Importantly, STING is decreased in islets of db/db mice and patients with T2D, suggesting a possible role of STING in β-cell dysfunction. Indeed, STING-βKO caused glucose intolerance due to impaired GSIS, indicating that STING is required for normal β-cell function. Islet transcriptome analysis showed that STING deficiency decreased expression of β-cell function–related genes, including Glut2, Kcnj11, and Abcc8, contributing to impaired GSIS. Mechanistically, the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and cleavage under targets and tagmentation (CUT&Tag) analyses suggested that Pax6 was the transcription factor that might be associated with defective GSIS in STING-βKO mice. Indeed, Pax6 messenger RNA and protein levels were down-regulated and its nuclear localization was lost in STING-βKO β-cells. Together, these data revealed a function of STING in the regulation of insulin secretion and established pathophysiological significance of fine-tuned STING within β-cells and insulin target tissues for maintaining glucose homeostasis.
Collapse
|
13
|
Harlan B, Park HG, Spektor R, Cummings B, Brenna JT, Soloway PD. Single-cell chromatin accessibility and lipid profiling reveals SCD1-dependent metabolic shift in adipocytes induced by bariatric surgery. PLoS One 2021; 16:e0261783. [PMID: 34972124 PMCID: PMC8719700 DOI: 10.1371/journal.pone.0261783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/09/2021] [Indexed: 11/23/2022] Open
Abstract
Obesity promotes type 2 diabetes and cardiometabolic pathologies. Vertical sleeve gastrectomy (VSG) is used to treat obesity resulting in long-term weight loss and health improvements that precede weight loss; however, the mechanisms underlying the immediate benefits remain incompletely understood. Because adipose plays a crucial role in energy homeostasis and utilization, we hypothesized that VSG exerts its influences, in part, by modulating adipose functional states. We applied single-cell ATAC sequencing and lipid profiling to inguinal and epididymal adipose depots from mice that received sham surgery or VSG. We observed depot-specific cellular composition and chromatin accessibility patterns that were altered by VSG. Specifically, accessibility at Scd1, a fatty acid desaturase, was substantially reduced after VSG in mature adipocytes of inguinal but not epididymal depots. This was accompanied by reduced accumulation of SCD1-produced unsaturated fatty acids. Given these findings and reports that reductions in Scd1 attenuate obesity and insulin resistance our results suggest VSG exerts its beneficial effects through an inguinal depot-specific reduction of SCD1 activity.
Collapse
Affiliation(s)
- Blaine Harlan
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Hui Gyu Park
- Dell Pediatric Research Institute, Department of Pediatrics, University of Texas at Austin, Austin, Texas, United States of America
| | - Roman Spektor
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Bethany Cummings
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - J. Thomas Brenna
- Dell Pediatric Research Institute, Department of Pediatrics, University of Texas at Austin, Austin, Texas, United States of America
- Division of Nutritional Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States of America
| | - Paul D. Soloway
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- Division of Nutritional Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
14
|
Wang Q, Zhang P, Cakir I, Mi L, Cone RD, Lin JD. Deletion of the Feeding-Induced Hepatokine TSK Ameliorates the Melanocortin Obesity Syndrome. Diabetes 2021; 70:2081-2091. [PMID: 34183373 PMCID: PMC8576423 DOI: 10.2337/db21-0161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/17/2021] [Indexed: 11/13/2022]
Abstract
Work in recent decades has established that metabolic hormones released by endocrine cells and diverse other cell types serve to regulate nutrient intake and energy homeostasis. Tsukushi (TSK) is a leucine-rich repeat-containing protein secreted primarily by the liver that exerts an inhibitory effect on brown fat sympathetic innervation and thermogenesis. Despite this, physiological regulation of TSK and the mechanisms underlying its effects on energy balance remain poorly understood. Here we show that hepatic expression and plasma concentrations of TSK are induced by feeding and regulated by melanocortin-4 receptor (MC4R) signaling. We generated TSK and MC4R-double-knockout mice to elucidate the nature of cross talk between TSK and the central regulatory circuit of energy balance. Remarkably, TSK inactivation restores energy balance, ameliorates hyperphagia, and improves metabolic health in MC4R-deficient mice. TSK ablation enhances thermogenic gene expression in brown fat, dampens obesity-association inflammation in the liver and adipose tissue, and protects MC4R-null mice from diet-induced nonalcoholic steatohepatitis. At the cellular level, TSK deficiency augments feeding-induced c-Fos expression in the paraventricular nucleus of the hypothalamus. These results illustrate physiological cross talk between TSK and the central regulatory circuit in maintaining energy balance and metabolic homeostasis.
Collapse
Affiliation(s)
- Qiuyu Wang
- Life Sciences Institute and Department of Cell & Developmental Biology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Peng Zhang
- Life Sciences Institute and Department of Cell & Developmental Biology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Isin Cakir
- Life Sciences Institute and Department of Molecular & Integrated Physiology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Lin Mi
- Life Sciences Institute and Department of Cell & Developmental Biology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Roger D Cone
- Life Sciences Institute and Department of Molecular & Integrated Physiology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, Michigan Medicine, University of Michigan, Ann Arbor, MI
| |
Collapse
|
15
|
Brandão BB, Poojari A, Rabiee A. Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential. Int J Mol Sci 2021; 22:5906. [PMID: 34072788 PMCID: PMC8198523 DOI: 10.3390/ijms22115906] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The concerning worldwide increase of obesity and chronic metabolic diseases, such as T2D, dyslipidemia, and cardiovascular disease, motivates further investigations into preventive and alternative therapeutic approaches. Over the past decade, there has been growing evidence that the formation and activation of thermogenic adipocytes (brown and beige) may serve as therapy to treat obesity and its associated diseases owing to its capacity to increase energy expenditure and to modulate circulating lipids and glucose levels. Thus, understanding the molecular mechanism of brown and beige adipocytes formation and activation will facilitate the development of strategies to combat metabolic disorders. Here, we provide a comprehensive overview of pathways and players involved in the development of brown and beige fat, as well as the role of thermogenic adipocytes in energy homeostasis and metabolism. Furthermore, we discuss the alterations in brown and beige adipose tissue function during obesity and explore the therapeutic potential of thermogenic activation to treat metabolic syndrome.
Collapse
Affiliation(s)
- Bruna B. Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Ankita Poojari
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| | - Atefeh Rabiee
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
16
|
Abstract
The mechanisms of epigenetic gene regulation-histone modifications, chromatin remodeling, DNA methylation, and noncoding RNA-use metabolites as enzymatic cofactors and substrates in reactions that allow chromatin formation, nucleotide biogenesis, transcription, RNA processing, and translation. Gene expression responds to demands from cellular processes that use specific metabolites and alters or maintains cellular metabolic status. However, the roles of metabolites-particularly nucleotides-as regulatory molecules in epigenetic regulation and biological processes remain largely unknown. Here we review the crosstalk between gene expression, nucleotide metabolism, and cellular processes, and explore the role of metabolism in epigenetics as a critical regulator of biological events.
Collapse
|
17
|
Shkodra B, Press AT, Vollrath A, Nischang I, Schubert S, Hoeppener S, Haas D, Enzensperger C, Lehmann M, Babic P, Benecke KJ, Traeger A, Bauer M, Schubert US. Formulation of Liver-Specific PLGA-DY-635 Nanoparticles Loaded with the Protein Kinase C Inhibitor Bisindolylmaleimide I. Pharmaceutics 2020; 12:pharmaceutics12111110. [PMID: 33218172 PMCID: PMC7698893 DOI: 10.3390/pharmaceutics12111110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Bisindolylmaleimide I (BIM-I) is a competitive pan protein kinase C inhibitor with anti-inflammatory and anti-metastatic properties, suggested to treat inflammatory diseases and various cancer entities. However, despite its therapeutic potential, BIM-I has two major drawbacks, i.e., it has a poor water solubility, and it binds the human ether-à-go-go-related gene (hERG) ion channels, potentially causing deadly arrhythmias. In this case, a targeted delivery of BIM-I is imperative to minimize peripheral side effects. To circumvent these drawbacks BIM-I was encapsulated into nanoparticles prepared from poly(lactic-co-glycolic acid) (PLGA) functionalized by the near-infrared dye DY-635. DY-635 served as an active targeting moiety since it selectively binds the OATP1B1 and OATP1B3 transporters that are highly expressed in liver and cancer cells. PLGA-DY-635 (BIM-I) nanoparticles were produced by nanoprecipitation and characterized using dynamic light scattering, analytical ultracentrifugation, and cryogenic transmission electron microscopy. Particle sizes were found to be in the range of 20 to 70 nm, while a difference in sizes between the drug-loaded and unloaded particles was observed by all analytical techniques. In vitro studies demonstrated that PLGA-DY-635 (BIM-I) NPs prevent the PKC activation efficiently, proving the efficacy of the inhibitor after its encapsulation, and suggesting that BIM-I is released from the PLGA-NPs. Ultimately, our results present a feasible formulation strategy that improved the cytotoxicity profile of BIM-I and showed a high cellular uptake in the liver as demonstrated in vivo by intravital microscopy investigations.
Collapse
Affiliation(s)
- Blerina Shkodra
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; (B.S.); (A.V.); (I.N.); (S.H.); (D.H.); (A.T.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
| | - Adrian T. Press
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.T.P.); (P.B.); (K.J.B.)
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Antje Vollrath
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; (B.S.); (A.V.); (I.N.); (S.H.); (D.H.); (A.T.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
| | - Ivo Nischang
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; (B.S.); (A.V.); (I.N.); (S.H.); (D.H.); (A.T.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
| | - Stephanie Schubert
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
- Institute of Pharmacy, Department of Pharmaceutical Technology and Biopharmacy, Friedrich Schiller University Jena, Lessingstrasse 8, 07743 Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; (B.S.); (A.V.); (I.N.); (S.H.); (D.H.); (A.T.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
| | - Dorothee Haas
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; (B.S.); (A.V.); (I.N.); (S.H.); (D.H.); (A.T.)
| | | | - Marc Lehmann
- SmartDyeLivery GmbH, Botzstrasse 5, 07743 Jena, Germany; (C.E.); (M.L.)
| | - Petra Babic
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.T.P.); (P.B.); (K.J.B.)
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Kay Jovana Benecke
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.T.P.); (P.B.); (K.J.B.)
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; (B.S.); (A.V.); (I.N.); (S.H.); (D.H.); (A.T.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
| | - Michael Bauer
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
- Department of Anesthesiology and Intensive Care Medicine, Nanophysiology Group, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (A.T.P.); (P.B.); (K.J.B.)
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Ulrich S. Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany; (B.S.); (A.V.); (I.N.); (S.H.); (D.H.); (A.T.)
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany; (S.S.); (M.B.)
- Correspondence: ; Tel.: +49-(0)-3641-9482-00
| |
Collapse
|