1
|
Loft A, Emont MP, Weinstock A, Divoux A, Ghosh A, Wagner A, Hertzel AV, Maniyadath B, Deplancke B, Liu B, Scheele C, Lumeng C, Ding C, Ma C, Wolfrum C, Strieder-Barboza C, Li C, Truong DD, Bernlohr DA, Stener-Victorin E, Kershaw EE, Yeger-Lotem E, Shamsi F, Hui HX, Camara H, Zhong J, Kalucka J, Ludwig JA, Semon JA, Jalkanen J, Whytock KL, Dumont KD, Sparks LM, Muir LA, Fang L, Massier L, Saraiva LR, Beyer MD, Jeschke MG, Mori MA, Boroni M, Walsh MJ, Patti ME, Lynes MD, Blüher M, Rydén M, Hamda N, Solimini NL, Mejhert N, Gao P, Gupta RK, Murphy R, Pirouzpanah S, Corvera S, Tang S, Das SK, Schmidt SF, Zhang T, Nelson TM, O'Sullivan TE, Efthymiou V, Wang W, Tong Y, Tseng YH, Mandrup S, Rosen ED. Towards a consensus atlas of human and mouse adipose tissue at single-cell resolution. Nat Metab 2025:10.1038/s42255-025-01296-9. [PMID: 40360756 DOI: 10.1038/s42255-025-01296-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025]
Abstract
Adipose tissue (AT) is a complex connective tissue with a high relative proportion of adipocytes, which are specialized cells with the ability to store lipids in large droplets. AT is found in multiple discrete depots throughout the body, where it serves as the primary repository for excess calories. In addition, AT has an important role in functions as diverse as insulation, immunity and regulation of metabolic homeostasis. The Human Cell Atlas Adipose Bionetwork was established to support the generation of single-cell atlases of human AT as well as the development of unified approaches and consensus for cell annotation. Here, we provide a first roadmap from this bionetwork, including our suggested cell annotations for humans and mice, with the aim of describing the state of the field and providing guidelines for the production, analysis, interpretation and presentation of AT single-cell data.
Collapse
Affiliation(s)
- Anne Loft
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark.
| | - Margo P Emont
- Section of Endocrinology, Diabetes and Metabolism, University of Chicago, Chicago, IL, USA.
| | - Ada Weinstock
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Adhideb Ghosh
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Allon Wagner
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, The University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Carey Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Changhai Ding
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Chenkai Ma
- Human Health, Health and Biosecurity, CSIRO, Canberra, Australian Capital Territory, Australia
| | - Christian Wolfrum
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Clarissa Strieder-Barboza
- Department of Veterinary Sciences, Texas Tech University, Lubbock, TX, USA
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, USA
| | - Congru Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Danh D Truong
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, The University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | | | - Erin E Kershaw
- Department of Medicine, Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, New York University, New York, NY, USA
- Departments of Cell Biology and Medicine, Grossman School of Medicine, New York University, New York, NY, USA
| | - Hannah X Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Henrique Camara
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jiawei Zhong
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Jutta Jalkanen
- Department of Medicine Huddinge (H7), Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Kyle D Dumont
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lindsey A Muir
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Lucas Massier
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Luis R Saraiva
- Sidra Medicine, Doha, Qatar
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Marc D Beyer
- Immunogenomics and Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Platform for Single Cell Genomics and Epigenomics (PRECISE), German Center for Neurodegenerative Diseases (DZNE) and University of Bonn and West German Genome Center (WGGC), Bonn, Germany
| | - Marc G Jeschke
- Centre for Burn Research, Hamilton Health Sciences Centre, Department of Surgery and Department of Biochemistry, McMaster University, Hamilton, Ontario, Canada
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
- Obesity and Comorbidities Research Center (OCRC), Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Martin J Walsh
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary-Elizabeth Patti
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Department of Medicine - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, Stockholm, Sweden
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | | | - Nicole L Solimini
- Department of Medical Oncology, Sarcoma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, C2-94, Karolinska University Hospital, Stockholm, Sweden
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Peng Gao
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Saeed Pirouzpanah
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Corvera
- University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Su'an Tang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Swapan K Das
- Department of Internal Medicine, Section on Endocrinology and Metabolism, Medical Center Boulevard, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Søren F Schmidt
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark
| | - Tao Zhang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore M Nelson
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Vissarion Efthymiou
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Wenjing Wang
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yihan Tong
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity (ATLAS), Department of Biochemistry and Molecular Biology, University of Southern Denmark (SDU), Odense, Denmark.
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025; 21:272-288. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
3
|
Wang S, Cui Y, Wang L, Feng C, Sun Y, Huo B, Jiang H, Zhao M, Tu Y, Wang Q, Yang Y, Zhang Q. Aregs-IGFBP3-mediated SMC-like cells apoptosis impairs beige adipocytes formation in aged mice. Mol Metab 2025; 95:102125. [PMID: 40118146 PMCID: PMC11985090 DOI: 10.1016/j.molmet.2025.102125] [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: 01/15/2025] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/23/2025] Open
Abstract
Aging is associated with a decline in the browning capacity of white adipose tissue (WAT), contributing to metabolic dysfunction. Beige adipocytes, which dissipate excess energy as heat, are a key feature of this process. In this study, we investigate the role of adipose stem and progenitor cells (ASPCs), specifically the Aregs (CD142+) subpopulation, in regulating beige adipocyte formation in aged mice under cold stimulation. Our findings reveal that Aregs significantly increase in the subcutaneous WAT (sWAT) of aged mice following cold exposure. We further demonstrate that Aregs secrete insulin-like growth factor binding protein 3 (IGFBP3), which appears to play a pivotal role in the cross-talk between adipogenesis-regulatory cells (Aregs) and smooth muscle cell-like (SMC-like) cells, thereby leading to the inhibition of beige adipocytes formation. Functional enrichment analysis highlighted the activation of TGFβ, MAPK and p53 signaling pathways in SMC-like cells, all of which are known to induce cell apoptosis and fibrosis. Moreover, IGFBP3 was found to interact with receptors and signaling molecules, including Egfr, Irf1 and Cdkn1a, in SMC-like cells, enhancing their apoptosis. Co-culture experiments confirmed that IGFBP3 significantly suppressed the formation of beige adipocytes, further corroborating its role in impairing browning. Overall, our study provides novel insights into the molecular mechanisms by which Aregs and IGFBP3 contribute to the age-related decline in WAT browning. These findings suggest potential therapeutic targets for reversing impaired WAT browning in aging and related metabolic disorders.
Collapse
Affiliation(s)
- Shifeng Wang
- Animal Zoology Department, Kunming Medical University, Kunming, 650000, China; School of Anesthesiology, Zunyi Medical University, Zunyi, 563000, China
| | - Yuanxu Cui
- Animal Zoology Department, Kunming Medical University, Kunming, 650000, China; Department of Emergency, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China; Science and Technology Achievement Transformation Center, Kunming Medical University, Kunming, 635000, China
| | - Limei Wang
- Animal Zoology Department, Kunming Medical University, Kunming, 650000, China
| | - Chun Feng
- Department of Otolaryngology, The First People's Hospital of Yunnan Province, Kunming, 650000, China
| | - Yifei Sun
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, 650000, China
| | - Bangyun Huo
- Department of Otolaryngology, The First People's Hospital of Yunnan Province, Kunming, 650000, China
| | - Honglu Jiang
- Department of Otolaryngology, The First People's Hospital of Yunnan Province, Kunming, 650000, China
| | - Mingyu Zhao
- The First School of Clinical Medicine, Kunming Medical University, Kunming, 650000, China
| | - Yingying Tu
- The First School of Clinical Medicine, Kunming Medical University, Kunming, 650000, China
| | - Qiyue Wang
- The First School of Clinical Medicine, Kunming Medical University, Kunming, 650000, China
| | - Yutao Yang
- The First School of Clinical Medicine, Kunming Medical University, Kunming, 650000, China
| | - Qiang Zhang
- Animal Zoology Department, Kunming Medical University, Kunming, 650000, China.
| |
Collapse
|
4
|
Zhang Z, Ni Z, Li T, Ning M, Gao C, Hu J, Han M, Yang J, Wu F, Chen L, Lu L, Wu Z, Ai H, Huang Y. Nine high-quality Anas genomes provide insights into Anas evolution and domestication. Cell Rep 2025; 44:115477. [PMID: 40173044 DOI: 10.1016/j.celrep.2025.115477] [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: 09/30/2024] [Revised: 12/18/2024] [Accepted: 03/06/2025] [Indexed: 04/04/2025] Open
Abstract
Evolutionary studies of wild and domestic organisms have yielded fascinating discoveries, while the species diversity and the domestication of ducks remain unclear. Here, we assembled eight chromosome-level Anas genomes, combined with the Pekin duck genome, to investigate Anas evolution and domestication. We found that, compared to autosomes, the Z chromosome was less affected by introgression and exhibited relatively stable local phylogenies. From the Z chromosome perspective, we proposed that the speciation of Anas platyrhynchos and Anas zonorhyncha was accompanied by continuous female-biased gene flow and remodeled duck domestication history. Moreover, we constructed an Anas pan-genome and identified several differentiated structural variations (SVs) between domestic and wild ducks. These SVs likely regulate their neighboring genes (i.e., GHR and FER), which represented the promising "domestication genes." Furthermore, a long terminal repeat (LTR) retrotransposon burst was found to have accelerated duck domestication, specifically contributing to functional shifts of the notable MITF and IGF2BP1 genes. These findings presented a live example for understanding animal evolutionary processes.
Collapse
Affiliation(s)
- Zhou Zhang
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China; National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang 330000, China
| | - Zijia Ni
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China
| | - Te Li
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China
| | - Mengfei Ning
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China
| | - Chuze Gao
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China
| | - Jiaxiang Hu
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China
| | - Mengying Han
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China
| | - Jiawen Yang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang 330000, China
| | - Fusheng Wu
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang 330000, China
| | - Li Chen
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310000, China
| | - Lizhi Lu
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310000, China
| | - Zhongzi Wu
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang 330000, China
| | - Huashui Ai
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang 330000, China
| | - Yinhua Huang
- State Key Laboratory for Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing 100000, China.
| |
Collapse
|
5
|
Wang HY, Peng XM, Yang M, Weng Y, Yang X, Zhan D, Ning Q, Luo XP, Chen Y. C5aR1-positive adipocytes mediate non-shivering thermogenesis in neonatal mice. iScience 2024; 27:111261. [PMID: 39758991 PMCID: PMC11700647 DOI: 10.1016/j.isci.2024.111261] [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: 04/08/2024] [Revised: 07/25/2024] [Accepted: 10/23/2024] [Indexed: 01/07/2025] Open
Abstract
Brown adipose tissue (BAT) plays an important role in maintaining body temperature in newborn mammals; however, its mechanisms remain poorly understood. Here, we report the identification of a special population of brown adipose tissue-derived stromal cells (ASCs) in neonatal mice that highly express CD45 and can be differentiated into adipocytes with lower thermogenic ability. These CD45+ adipocytes also characteristically contained complement C5a receptor 1(C5aR1) on the cell membrane. C5ar1 deficiency in BAT resulted in an apparent immaturity of adipocytes and cold intolerance in neonatal mice. Mechanistically, loss of C5aR1 in these CD45+ brown adipocytes caused an increase in the secretion of plate factor four (PF4) from these cells, suppressing the maturity of neighboring brown adipocytes. Overall, our results indicated that the accumulation of C5aR1 positive brown adipocyte in neonatal BAT is essential for thermoregulation in newborn mice, which unveiled the regulatory mechanism of BAT-mediated thermogenesis in newborns.
Collapse
Affiliation(s)
- Huan-Yu Wang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue-Min Peng
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Yang
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Weng
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Xi Yang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Di Zhan
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Qin Ning
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Ping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Pediatric Genetic Metabolic and Endocrine Rare Diseases, Wuhan 430030, China
| | - Yong Chen
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
6
|
Wang Y, Thottappillil N, Gomez-Salazar M, Tower RJ, Qin Q, Del Rosario Alvia IC, Xu M, Cherief M, Cheng R, Archer M, Arondekar S, Reddy S, Broderick K, Péault B, James AW. Integrated transcriptomics of human blood vessels defines a spatially controlled niche for early mesenchymal progenitor cells. Dev Cell 2024; 59:2687-2703.e6. [PMID: 39025061 PMCID: PMC11496018 DOI: 10.1016/j.devcel.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/28/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024]
Abstract
Human blood vessel walls show concentric layers, with the outermost tunica adventitia harboring mesenchymal progenitor cells. These progenitor cells maintain vessel homeostasis and provide a robust cell source for cell-based therapies. However, human adventitial stem cell niche has not been studied in detail. Here, using spatial and single-cell transcriptomics, we characterized the phenotype, potential, and microanatomic distribution of human perivascular progenitors. Initially, spatial transcriptomics identified heterogeneity between perivascular layers of arteries and veins and delineated the tunica adventitia into inner and outer layers. From this spatial atlas, we inferred a hierarchy of mesenchymal progenitors dictated by a more primitive cell with a high surface expression of CD201 (PROCR). When isolated from humans and mice, CD201Low expression typified a mesodermal committed subset with higher osteogenesis and less proliferation than CD201High cells, with a downstream effect on canonical Wnt signaling through DACT2. CD201Low cells also displayed high translational potential for bone tissue generation.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | - Robert J Tower
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ray Cheng
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shreya Arondekar
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sashank Reddy
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kristen Broderick
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bruno Péault
- Department of Orthopedic Surgery and Orthopedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Mo YY, Han YX, Xu SN, Jiang HL, Wu HX, Cai JM, Li L, Bu YH, Xiao F, Liang HD, Wen Y, Liu YZ, Yin YL, Zhou HD. Adipose Tissue Plasticity: A Comprehensive Definition and Multidimensional Insight. Biomolecules 2024; 14:1223. [PMID: 39456156 PMCID: PMC11505740 DOI: 10.3390/biom14101223] [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: 08/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Adipose tissue is composed of adipocytes, stromal vascular fraction, nerves, surrounding immune cells, and the extracellular matrix. Under various physiological or pathological conditions, adipose tissue shifts cellular composition, lipid storage, and organelle dynamics to respond to the stress; this remodeling is called "adipose tissue plasticity". Adipose tissue plasticity includes changes in the size, species, number, lipid storage capacity, and differentiation function of adipocytes, as well as alterations in the distribution and cellular composition of adipose tissue. This plasticity has a major role in growth, obesity, organismal protection, and internal environmental homeostasis. Moreover, certain thresholds exist for this plasticity with significant individualized differences. Here, we comprehensively elaborate on the specific connotation of adipose tissue plasticity and the relationship between this plasticity and the development of many diseases. Meanwhile, we summarize possible strategies for treating obesity in response to adipose tissue plasticity, intending to provide new insights into the dynamic changes in adipose tissue and contribute new ideas to relevant clinical problems.
Collapse
Affiliation(s)
- Yu-Yao Mo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Xin Han
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Shi-Na Xu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Jun-Min Cai
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yan-Hong Bu
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha 410012, China;
| | - Fen Xiao
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Han-Dan Liang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Ying Wen
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Ze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
| | - Yu-Long Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| |
Collapse
|
8
|
Ferrero R, Rainer PY, Rumpler M, Russeil J, Zachara M, Pezoldt J, van Mierlo G, Gardeux V, Saelens W, Alpern D, Favre L, Vionnet N, Mantziari S, Zingg T, Pitteloud N, Suter M, Matter M, Schlaudraff KU, Canto C, Deplancke B. A human omentum-specific mesothelial-like stromal population inhibits adipogenesis through IGFBP2 secretion. Cell Metab 2024; 36:1566-1585.e9. [PMID: 38729152 DOI: 10.1016/j.cmet.2024.04.017] [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: 05/12/2023] [Revised: 12/22/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024]
Abstract
Adipose tissue plasticity is orchestrated by molecularly and functionally diverse cells within the stromal vascular fraction (SVF). Although several mouse and human adipose SVF cellular subpopulations have by now been identified, we still lack an understanding of the cellular and functional variability of adipose stem and progenitor cell (ASPC) populations across human fat depots. To address this, we performed single-cell and bulk RNA sequencing (RNA-seq) analyses of >30 SVF/Lin- samples across four human adipose depots, revealing two ubiquitous human ASPC (hASPC) subpopulations with distinct proliferative and adipogenic properties but also depot- and BMI-dependent proportions. Furthermore, we identified an omental-specific, high IGFBP2-expressing stromal population that transitions between mesothelial and mesenchymal cell states and inhibits hASPC adipogenesis through IGFBP2 secretion. Our analyses highlight the molecular and cellular uniqueness of different adipose niches, while our discovery of an anti-adipogenic IGFBP2+ omental-specific population provides a new rationale for the biomedically relevant, limited adipogenic capacity of omental hASPCs.
Collapse
Affiliation(s)
- Radiana Ferrero
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Pernille Yde Rainer
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Marie Rumpler
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Julie Russeil
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Magda Zachara
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Joern Pezoldt
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Guido van Mierlo
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Vincent Gardeux
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Wouter Saelens
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Lucie Favre
- Department of Endocrinology, Diabetology and Metabolism, University Hospital of Lausanne (CHUV), 1011 Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Nathalie Vionnet
- Department of Endocrinology, Diabetology and Metabolism, University Hospital of Lausanne (CHUV), 1011 Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Styliani Mantziari
- Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne 1011, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Tobias Zingg
- Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne 1011, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Nelly Pitteloud
- Department of Endocrinology, Diabetology and Metabolism, University Hospital of Lausanne (CHUV), 1011 Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Michel Suter
- Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne 1011, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Maurice Matter
- Department of Visceral Surgery, University Hospital of Lausanne (CHUV), Lausanne 1011, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | | | - Carles Canto
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| |
Collapse
|
9
|
Wang J, Gu Y, Sun Y, Qiao Q, Huang X, Yang K, Bai Y. Adipogenic differentiation effect of human periodontal ligament stem cell initial cell density on autologous cells and human bone marrow stromal cells. Cell Biochem Funct 2024; 42:e4069. [PMID: 38940455 DOI: 10.1002/cbf.4069] [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: 12/08/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024]
Abstract
Stem cells demonstrate differentiation and regulatory functions. In this discussion, we will explore the impacts of cell culture density on stem cell proliferation, adipogenesis, and regulatory abilities. This study aimed to investigate the impact of the initial culture density of human periodontal ligament stem cells (hPDLSCs) on the adipogenic differentiation of autologous cells. Our findings indicate that the proliferation rate of hPDLSCs increased with increasing initial cell density (0.5-8 × 104 cells/cm2). After adipogenic differentiation induced by different initial cell densities of hPDLSC, we found that the mean adipose concentration and the expression levels of lipoprotein lipase (LPL), CCAAT/enhancer binding protein α (CEBPα), and peroxisome proliferator-activated receptor γ (PPAR-γ) genes all increased with increasing cell density. To investigate the regulatory role of hPDLSCs in the adipogenic differentiation of other cells, we used secreted exocrine vesicles derived from hPDLSCs cultivated at different initial cell densities of 50 μg/mL to induce the adipogenic differentiation of human bone marrow stromal cells. We also found that the mean adipose concentration and expression of LPL, CEBPα, and PPARγ genes increased with increasing cell density, with an optimal culture density of 8 × 104 cells/cm2. This study provides a foundation for the application of adipogenic differentiation in stem cells.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yingzhi Gu
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yaxi Sun
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Qingchen Qiao
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Huang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Yi X, Feng M, Zhu J, Yu H, He Z, Zhang Z, Zhao T, Zhang Q, Pang W. Adipocyte Progenitor Pools Composition and Cellular Niches Affect Adipogenesis Divergence in Porcine Subcutaneous and Intramuscular Fat. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38848240 DOI: 10.1021/acs.jafc.4c01044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Intramuscular fat (IMF) contributed positively to pork quality, whereas subcutaneous fat (SCF) was often considered to be a detrimental factor impacting growth and carcass traits. Reducing SCF while maintaining optimal IMF levels requires a thorough understanding of the adipogenic differences between these two adipose depots. Our study explored the differences in adipogenesis between porcine IMF and SCF, and the results showed that subcutaneous adipocytes (SCAs) demonstrate a greater potential for adipogenic differentiation, both in vivo and in vitro. Lipidomic and transcriptomic analyses suggested that intramuscular adipocytes (IMAs) are more inclined to biosynthesize unsaturated fatty acids. Furthermore, single-cell RNA sequencing (scRNA-seq) was employed to dissect the intrinsic and microenvironmental discrepancies in adipogenesis between porcine IMF and SCF. Comparative analysis indicated that SCF was enriched with preadipocytes, exhibiting an enhanced adipogenic potential, while IMF was characterized by a higher abundance of stem cells. Furthermore, coculture analyses of porcine intramuscular adipogenic cells and myogenetic cells indicated that the niche of IMAs inhibited its adipogenic differentiation. Cell communication analysis identified 160 ligand-receptor pairs and channels between adipogenic and myogenetic cells in IMF. Collectively, our study elucidated two intrinsic and microenvironmental novel mechanisms underpinning the divergence in adipogenesis between porcine SCF and IMF.
Collapse
Affiliation(s)
- Xudong Yi
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ming Feng
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jiahua Zhu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - He Yu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhaozhao He
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ziyi Zhang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Tiantian Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Que Zhang
- Department of Animal Science and Technology, Shandong Vocational Animal Science and Veterinary College, Weifang, Shandong 261061, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
11
|
Yang Z, Chen F, Zhang Y, Ou M, Tan P, Xu X, Li Q, Zhou S. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm (Beijing) 2024; 5:e560. [PMID: 38812572 PMCID: PMC11134193 DOI: 10.1002/mco2.560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024] Open
Abstract
White adipose tissue is not only a highly heterogeneous organ containing various cells, such as adipocytes, adipose stem and progenitor cells, and immune cells, but also an endocrine organ that is highly important for regulating metabolic and immune homeostasis. In individuals with obesity, dynamic cellular changes in adipose tissue result in phenotypic switching and adipose tissue dysfunction, including pathological expansion, WAT fibrosis, immune cell infiltration, endoplasmic reticulum stress, and ectopic lipid accumulation, ultimately leading to chronic low-grade inflammation and insulin resistance. Recently, many distinct subpopulations of adipose tissue have been identified, providing new insights into the potential mechanisms of adipose dysfunction in individuals with obesity. Therefore, targeting white adipose tissue as a therapeutic agent for treating obesity and obesity-related metabolic diseases is of great scientific interest. Here, we provide an overview of white adipose tissue remodeling in individuals with obesity including cellular changes and discuss the underlying regulatory mechanisms of white adipose tissue metabolic dysfunction. Currently, various studies have uncovered promising targets and strategies for obesity treatment. We also outline the potential therapeutic signaling pathways of targeting adipose tissue and summarize existing therapeutic strategies for antiobesity treatment including pharmacological approaches, lifestyle interventions, and novel therapies.
Collapse
Affiliation(s)
- Zi‐Han Yang
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang‐Zhou Chen
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Xiang Zhang
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min‐Yi Ou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Poh‐Ching Tan
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue‐Wen Xu
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qing‐Feng Li
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuang‐Bai Zhou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
12
|
Zhang J, Jiang J, Liu H, Wang S, Ke K, Liu S, Jiang Y, Liu L, Gao X, He B, Su Y. BMP9 induces osteogenic differentiation through up-regulating LGR4 via the mTORC1/Stat3 pathway in mesenchymal stem cells. Genes Dis 2024; 11:101075. [PMID: 38292169 PMCID: PMC10825279 DOI: 10.1016/j.gendis.2023.101075] [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: 03/25/2023] [Revised: 06/21/2023] [Accepted: 07/24/2023] [Indexed: 02/01/2024] Open
Abstract
Bone defects and non-union are prevalent in clinical orthopedy, and the outcomes of current treatments are often suboptimal. Bone tissue engineering offers a promising approach to treating these conditions effectively. Bone morphogenetic protein 9 (BMP9) can commit mesenchymal stem cells to osteogenic lineage, and a knowledge of the underlying mechanisms may help advance the field of bone tissue engineering. Leucine-rich repeats containing G protein-coupled receptor 4 (LGR4), a member of G protein-coupled receptors, is essential for modulating bone development. This study is aimed at investigating the impact of LGR4 on BMP9-induced osteogenesis in mesenchymal stem cells as well as the underlying mechanisms. Bone marrow stromal cells from BMP9-knockout mice exhibited diminished LGR4 expression, and exogenous LGR4 clearly restored the impaired osteogenic potency of the bone marrow stromal cells. Furthermore, LGR4 expression was increased by BMP9 in C3H10T1/2 cells. LGR4 augmented the benefits of BMP9-induced osteogenic markers and bone formation, whereas LGR4 inhibition restricted these effects. Meanwhile, the BMP9-induced lipogenic markers were increased by LGR4 inhibition. The protein levels of Raptor and p-Stat3 were elevated by BMP9. Raptor knockdown or p-Stat3 suppression attenuated the osteoblastic markers and LGR4 expression brought on by BMP9. LGR4 significantly reversed the blocking effect of Raptor knockdown or p-Stat3 suppression on the BMP9-induced osteoblastic markers. Raptor interacts with p-Stat3, and p-Stat3 activates the LGR4 promoter activity. In conclusion, LGR4 boosts BMP9 osteoblastic potency in mesenchymal stem cells, and BMP9 may up-regulate LGR4 via the mTORC1/Stat3 signal activation.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Jinhai Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Hang Liu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Orthopedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shiyu Wang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Kaixin Ke
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Siyuan Liu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Orthopedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yue Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Lu Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Xiang Gao
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Orthopedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Baicheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Yuxi Su
- Orthopedics Department, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Jiangxi Hospital Affiliated Children’s Hospital of Chongqing Medical University, Jiangxi 330000, China
- National Clinical Research Center for Child Health and Disorders, China
| |
Collapse
|
13
|
Navarro-Perez J, Carobbio S. Adipose tissue-derived stem cells, in vivo and in vitro models for metabolic diseases. Biochem Pharmacol 2024; 222:116108. [PMID: 38438053 DOI: 10.1016/j.bcp.2024.116108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
The primary role of adipose tissue stem cells (ADSCs) is to support the function and homeostasis of adipose tissue in physiological and pathophysiological conditions. However, when ADSCs become dysfunctional in diseases such as obesity and cancer, they become impaired, undergo signalling changes, and their epigenome is altered, which can have a dramatic effect on human health. In more recent years, the therapeutic potential of ADSCs in regenerative medicine, wound healing, and for treating conditions such as cancer and metabolic diseases has been extensively investigated with very promising results. ADSCs have also been used to generate two-dimensional (2D) and three-dimensional (3D) cellular and in vivo models to study adipose tissue biology and function as well as intracellular communication. Characterising the biology and function of ADSCs, how it is altered in health and disease, and its therapeutic potential and uses in cellular models is key for designing intervention strategies for complex metabolic diseases and cancer.
Collapse
|
14
|
Wu D, Khan FA, Zhang K, Pandupuspitasari NS, Negara W, Guan K, Sun F, Huang C. Retinoic acid signaling in development and differentiation commitment and its regulatory topology. Chem Biol Interact 2024; 387:110773. [PMID: 37977248 DOI: 10.1016/j.cbi.2023.110773] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Retinoic acid (RA), the derivative of vitamin A/retinol, is a signaling molecule with important implications in health and disease. It is a well-known developmental morphogen that functions mainly through the transcriptional activity of nuclear RA receptors (RARs) and, uncommonly, through other nuclear receptors, including peroxisome proliferator-activated receptors. Intracellular RA is under spatiotemporally fine-tuned regulation by synthesis and degradation processes catalyzed by retinaldehyde dehydrogenases and P450 family enzymes, respectively. In addition to dictating the transcription architecture, RA also impinges on cell functioning through non-genomic mechanisms independent of RAR transcriptional activity. Although RA-based differentiation therapy has achieved impressive success in the treatment of hematologic malignancies, RA also has pro-tumor activity. Here, we highlight the relevance of RA signaling in cell-fate determination, neurogenesis, visual function, inflammatory responses and gametogenesis commitment. Genetic and post-translational modifications of RAR are also discussed. A better understanding of RA signaling will foster the development of precision medicine to improve the defects caused by deregulated RA signaling.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | | | - Windu Negara
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
15
|
Shan B, Barker CS, Theraulaz H, Zhang X, Ping Y, Gupta RK, Shao M, Wu Y. Protocol for quantitative proteomic analysis of heterogeneous adipose tissue-residing progenitor subpopulations in mice. STAR Protoc 2023; 4:102676. [PMID: 38048219 PMCID: PMC10730372 DOI: 10.1016/j.xpro.2023.102676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/15/2023] [Accepted: 10/06/2023] [Indexed: 12/06/2023] Open
Abstract
Recent studies have revealed cellular heterogeneity of mesenchymal stromal cells and immune cells in adipose tissue and emphasized the need for quantitative analysis of small numbers of functionally distinct cells using state-of-the-art "omics" technologies. Here, we present an optimized protocol for precise protein quantification from minute amounts of samples. We describe steps for isolation of mouse adipose progenitor cells, proteomics sample preparation, mass spectrometry measurement, and computational analysis. This protocol can be adapted to other samples with limited amounts. For complete details on the use and execution of this protocol, please refer to Shan et al. (2022).1.
Collapse
Affiliation(s)
- Bo Shan
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China.
| | - Clive S Barker
- YCI Laboratory for Next-Generation Proteomics, RIKEN Center of Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Harry Theraulaz
- Chemical Biology Mass Spectrometry (ChemBioMS) Platform, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland
| | - Xiaoli Zhang
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Yan Ping
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China; Cancer Center, Zhejiang University, Hangzhou, China
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Mengle Shao
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
| | - Yibo Wu
- YCI Laboratory for Next-Generation Proteomics, RIKEN Center of Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Chemical Biology Mass Spectrometry (ChemBioMS) Platform, Faculty of Science, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
16
|
Couchet M, Gao H, Klingelhuber F, Jalkanen J, De Castro Barbosa T, Omar-Hmeadi M, Massier L, Krahmer N, Mejhert N, Rydén M. Adipogenic characterization of immortalized CD55 + progenitor cells from human white adipose tissue. Adipocyte 2023:2283213. [PMID: 37982546 DOI: 10.1080/21623945.2023.2283213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/08/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Mature adipocytes are notoriously difficult to study ex vivo and alternative cell culture systems have therefore been developed. One of the most common models are human adipose progenitor cells (hAPCs). Unfortunately, these display replicative senescence after prolonged culture conditions, which limits their use in mechanistic studies. METHODS Herein, we knocked in human telomerase reverse transcriptase (TERT) into the AAVS1 locus of CD55+ hAPCs derived from abdominal subcutaneous adipose tissue and characterized the cells before and after differentiation into adipocytes. RESULTS Immortalized TERT-hAPCs retained proliferative and adipogenic capacities comparable to those of early-passage wild type hAPCs for > 80 passages. In line with this, our integrative transcriptomic and proteomic analyses revealed that TERT-hAPCs displayed robust adipocyte expression profiles in comparison to wild type hAPCs. This was confirmed by functional analyses of lipid turnover where TERT-hAPCs exhibited pronounced responses to insulin and pro-lipolytic stimuli such as isoprenaline, dibutyrul cAMP and tumour necrosis factor alpha. In addition, TERT-hAPCs could be readily cultured in both standard 2D and 3D-cultures and proteomic analyses revealed that the spheroid culture conditions improved adipogenesis. CONCLUSION Through descriptive and functional studies, we demonstrate that immortalization of human CD55+ hAPCs is feasible and results in cells with stable proliferative and adipogenic capacities over multiple passages. As these cells are cryopreservable, they provide the additional advantage over primary cells of allowing repeated studies in both 2D and 3D model systems with the same genetic background. (234/250).
Collapse
Affiliation(s)
- Morgane Couchet
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Felix Klingelhuber
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany
- German Center forDiabetes Research, Neuherberg, Germany
| | - Jutta Jalkanen
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | | | | | - Lucas Massier
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg, Germany
- German Center forDiabetes Research, Neuherberg, Germany
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
He M, Li Z, Tung VSK, Pan M, Han X, Evgrafov O, Jiang XC. Inhibiting Phosphatidylcholine Remodeling in Adipose Tissue Increases Insulin Sensitivity. Diabetes 2023; 72:1547-1559. [PMID: 37625119 PMCID: PMC10588299 DOI: 10.2337/db23-0317] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Cell membrane phosphatidylcholine (PC) composition is regulated by lysophosphatidylcholine acyltransferase (LPCAT); changes in membrane PC saturation are implicated in metabolic disorders. Here, we identified LPCAT3 as the major isoform of LPCAT in adipose tissue and created adipocyte-specific Lpcat3-knockout mice to study adipose tissue lipid metabolism. Transcriptome sequencing and plasma adipokine profiling were used to investigate how LPCAT3 regulates adipose tissue insulin signaling. LPCAT3 deficiency reduced polyunsaturated PCs in adipocyte plasma membranes, increasing insulin sensitivity. LPCAT3 deficiency influenced membrane lipid rafts, which activated insulin receptors and AKT in adipose tissue, and attenuated diet-induced insulin resistance. Conversely, higher LPCAT3 activity in adipose tissue from ob/ob, db/db, and high-fat diet-fed mice reduced insulin signaling. Adding polyunsaturated PCs to mature human or mouse adipocytes in vitro worsened insulin signaling. We suggest that targeting LPCAT3 in adipose tissue to manipulate membrane phospholipid saturation is a new strategy to treat insulin resistance. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mulin He
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Zhiqiang Li
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Victoria Sook Keng Tung
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Meixia Pan
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xianlin Han
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Oleg Evgrafov
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Xian-Cheng Jiang
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
- Molecular and Cellular Cardiology Program, Veterans Affairs New York Harbor Healthcare System, New York, NY
| |
Collapse
|
18
|
Fleming SJ, Chaffin MD, Arduini A, Akkad AD, Banks E, Marioni JC, Philippakis AA, Ellinor PT, Babadi M. Unsupervised removal of systematic background noise from droplet-based single-cell experiments using CellBender. Nat Methods 2023; 20:1323-1335. [PMID: 37550580 DOI: 10.1038/s41592-023-01943-7] [Citation(s) in RCA: 212] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 06/13/2023] [Indexed: 08/09/2023]
Abstract
Droplet-based single-cell assays, including single-cell RNA sequencing (scRNA-seq), single-nucleus RNA sequencing (snRNA-seq) and cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq), generate considerable background noise counts, the hallmark of which is nonzero counts in cell-free droplets and off-target gene expression in unexpected cell types. Such systematic background noise can lead to batch effects and spurious differential gene expression results. Here we develop a deep generative model based on the phenomenology of noise generation in droplet-based assays. The proposed model accurately distinguishes cell-containing droplets from cell-free droplets, learns the background noise profile and provides noise-free quantification in an end-to-end fashion. We implement this approach in the scalable and robust open-source software package CellBender. Analysis of simulated data demonstrates that CellBender operates near the theoretically optimal denoising limit. Extensive evaluations using real datasets and experimental benchmarks highlight enhanced concordance between droplet-based single-cell data and established gene expression patterns, while the learned background noise profile provides evidence of degraded or uncaptured cell types.
Collapse
Affiliation(s)
- Stephen J Fleming
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Precision Cardiology Laboratory (PCL), Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Mark D Chaffin
- Precision Cardiology Laboratory (PCL), Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alessandro Arduini
- Precision Cardiology Laboratory (PCL), Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Bayer US, LLC, Cambridge, MA, USA
| | - Amer-Denis Akkad
- Precision Cardiology Laboratory (PCL), Bayer US, LLC, Cambridge, MA, USA
| | - Eric Banks
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John C Marioni
- Wellcome Sanger Institute, Hinxton, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
| | | | - Patrick T Ellinor
- Precision Cardiology Laboratory (PCL), Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Mehrtash Babadi
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Precision Cardiology Laboratory (PCL), Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
19
|
Abstract
Adipose tissue exhibits a remarkable capacity to expand, contract, and remodel in response to changes in physiological and environmental conditions. Here, we describe recent advances in our understanding of how functionally distinct tissue-resident mesenchymal stromal cell subpopulations orchestrate several aspects of physiological and pathophysiological adipose tissue remodeling, with a particular focus on the adaptations that occur in response to changes in energy surplus and environmental temperature. The study of adipose tissue remodeling provides a vehicle to understand the functional diversity of stromal cells and offers a lens through which several generalizable aspects of tissue reorganization can be readily observed.
Collapse
Affiliation(s)
- Jessica Cannavino
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| |
Collapse
|
20
|
Dewal RS, Wolfrum C. Master of disguise: deconvoluting adipose tissue heterogeneity and its impact on metabolic health. Curr Opin Genet Dev 2023; 81:102085. [PMID: 37421902 DOI: 10.1016/j.gde.2023.102085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/24/2023] [Accepted: 06/13/2023] [Indexed: 07/10/2023]
Abstract
Adipose tissue in its different forms: white, brown, and beige, while essential in day-to-day bodily functions, leads to several disorders when present in overabundance, including obesity and type-2 diabetes. Adipose tissue function/dysfunction is largely mediated by the diversity of its cell composition, within adipocytes and cells in its stromal fraction. Owing to its heterogeneous nature, recent studies have focused on intercalating the effects of cellular diversity with adipose tissue function, particularly by employing sequencing technologies. In this review, we highlight the recent advances in utilizing single-cell and single-nuclei RNA sequencing technologies to discover novel adipose tissue cell types or subtypes, and to determine their role in mediating tissue, as well as whole-body metabolism and function.
Collapse
Affiliation(s)
- Revati S Dewal
- Laboratory of Translational Nutritional Biology, Department of Health Sciences and Technology, ETH Zurich, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland. https://twitter.com/@revadewa3
| | - Christian Wolfrum
- Laboratory of Translational Nutritional Biology, Department of Health Sciences and Technology, ETH Zurich, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland.
| |
Collapse
|
21
|
Liu SQ, Chen DY, Li B, Gao ZJ, Feng HF, Yu X, Liu Z, Wang Y, Li WG, Sun S, Sun SR, Wu Q. Single-cell analysis of white adipose tissue reveals the tumor-promoting adipocyte subtypes. J Transl Med 2023; 21:470. [PMID: 37454080 PMCID: PMC10349475 DOI: 10.1186/s12967-023-04256-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND The tumor-adipose microenvironment (TAME) is characterized by the enrichment of adipocytes, and is considered a special ecosystem that supports cancer progression. However, the heterogeneity and diversity of adipocytes in TAME remains poorly understood. METHODS We conducted a single-cell RNA sequencing analysis of adipocytes in mouse and human white adipose tissue (WAT). We analyzed several adipocyte subtypes to evaluate their relationship and potential as prognostic factors for overall survival (OS). The potential drugs are screened by using bioinformatics methods. The tumor-promoting effects of a typical adipocyte subtype in breast cancer are validated by performing in vitro functional assays and immunohistochemistry (IHC) in clinical samples. RESULTS We profiled a comprehensive single-cell atlas of adipocyte in mouse and human WAT and described their characteristics, origins, development, functions and interactions with immune cells. Several cancer-associated adipocyte subtypes, namely DPP4+ adipocytes in visceral adipose and ADIPOQ+ adipocytes in subcutaneous adipose, are identified. We found that high levels of these subtypes are associated with unfavorable outcomes in four typical adipose-associated cancers. Some potential drugs including Trametinib, Selumetinib and Ulixertinib are discovered. Emphatically, knockdown of adiponectin receptor 1 (AdipoR1) and AdipoR2 impaired the proliferation and invasion of breast cancer cells. Patients with AdipoR2-high breast cancer display significantly shorter relapse-free survival (RFS) than those with AdipoR2-low breast cancer. CONCLUSION Our results provide a novel understanding of TAME at the single-cell level. Based on our findings, several adipocyte subtypes have negative impact on prognosis. These cancer-associated adipocytes may serve as key prognostic predictor and potential targets for treatment in the future.
Collapse
Affiliation(s)
- Si-Qing Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Ding-Yuan Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhi-Jie Gao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Hong-Fang Feng
- Department of Breast and Thyroid Surgery, Huangshi Central Hospital, Hubei Polytechnic University, Huangshi, Hubei, People's Republic of China
| | - Xin Yu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhou Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yuan Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Wen-Ge Li
- Department of Oncology, Shanghai Artemed Hospital, Shanghai, People's Republic of China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
22
|
Sun L, Zhang X, Wu S, Liu Y, Guerrero-Juarez CF, Liu W, Huang J, Yao Q, Yin M, Li J, Ramos R, Liao Y, Wu R, Xia T, Zhang X, Yang Y, Li F, Heng S, Zhang W, Yang M, Tzeng CM, Ji C, Plikus MV, Gallo RL, Zhang LJ. Dynamic interplay between IL-1 and WNT pathways in regulating dermal adipocyte lineage cells during skin development and wound regeneration. Cell Rep 2023; 42:112647. [PMID: 37330908 PMCID: PMC10765379 DOI: 10.1016/j.celrep.2023.112647] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023] Open
Abstract
Dermal adipocyte lineage cells are highly plastic and can undergo reversible differentiation and dedifferentiation in response to various stimuli. Using single-cell RNA sequencing of developing or wounded mouse skin, we classify dermal fibroblasts (dFBs) into distinct non-adipogenic and adipogenic cell states. Cell differentiation trajectory analyses identify IL-1-NF-κB and WNT-β-catenin as top signaling pathways that positively and negatively associate with adipogenesis, respectively. Upon wounding, activation of adipocyte progenitors and wound-induced adipogenesis are mediated in part by neutrophils through the IL-1R-NF-κB-CREB signaling axis. In contrast, WNT activation, by WNT ligand and/or ablation of Gsk3, inhibits the adipogenic potential of dFBs but promotes lipolysis and dedifferentiation of mature adipocytes, contributing to myofibroblast formation. Finally, sustained WNT activation and inhibition of adipogenesis is seen in human keloids. These data reveal molecular mechanisms underlying the plasticity of dermal adipocyte lineage cells, defining potential therapeutic targets for defective wound healing and scar formation.
Collapse
Affiliation(s)
- Lixiang Sun
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaowei Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shuai Wu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Youxi Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | | | - Wenjie Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jinwen Huang
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qian Yao
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Meimei Yin
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiacheng Li
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yanhang Liao
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Rundong Wu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Tian Xia
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xinyuan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yichun Yang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shujun Heng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wenlu Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Minggang Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 350005, China
| | - Chi-Meng Tzeng
- Translation Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Chao Ji
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ling-Juan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
23
|
Navarro-Perez J, Vidal-Puig A, Carobbio S. Recent developments in adipose tissue-secreted factors and their target organs. Curr Opin Genet Dev 2023; 80:102046. [PMID: 37099831 DOI: 10.1016/j.gde.2023.102046] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/12/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023]
Abstract
The white adipose tissue's primary roles are to store and mobilise energy, which is very different from the brown adipose tissue's function of using fuel to generate heat and maintain the body temperature. The adipose tissues (ATs), co-ordinately with the other organs, sense energetic demands and inform of their reserves before embarking on energetically demanding physiological functions. It is not surprising that ATs exhibit highly integrated regulatory mechanisms mediated by a diversified secretome, including adipokines, lipokines, metabolites and a repertoire of extracellular miRNAs that contribute to integrating the function of the AT niche and connect the AT through paracrine and endocrine effects with the whole organism. Characterising the adipose secretome, its changes in health and disease, regulation by ageing and gender and their contribution to energy homoeostasis is necessary to optimise its use for personalised strategies to prevent or reverse metabolic diseases.
Collapse
Affiliation(s)
- Jaime Navarro-Perez
- Centro de Investigacion Principe Felipe, Valencia, Spain. https://twitter.com/@JaimeNavarroPr1
| | - Antonio Vidal-Puig
- Centro de Investigacion Principe Felipe, Valencia, Spain; Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | | |
Collapse
|
24
|
Yang Loureiro Z, Joyce S, DeSouza T, Solivan-Rivera J, Desai A, Skritakis P, Yang Q, Ziegler R, Zhong D, Nguyen TT, MacDougald OA, Corvera S. Wnt signaling preserves progenitor cell multipotency during adipose tissue development. Nat Metab 2023; 5:1014-1028. [PMID: 37337125 PMCID: PMC10290956 DOI: 10.1038/s42255-023-00813-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 04/25/2023] [Indexed: 06/21/2023]
Abstract
Mesenchymal stem/progenitor cells are essential for tissue development and repair throughout life, but how they are maintained under chronic differentiation pressure is not known. Using single-cell transcriptomics of human progenitor cells we find that adipose differentiation stimuli elicit two cellular trajectories: one toward mature adipocytes and another toward a pool of non-differentiated cells that maintain progenitor characteristics. These cells are induced by transient Wnt pathway activation and express numerous extracellular matrix genes and are therefore named structural Wnt-regulated adipose tissue cells. We find that the genetic signature of structural Wnt-regulated adipose tissue cells is present in adult human adipose tissue and adipose tissue developed from human progenitor cells in mice. Our results suggest a mechanism whereby adipose differentiation occurs concurrently with the maintenance of a mesenchymal progenitor cell pool, ensuring tissue development, repair and appropriate metabolic control over the lifetime.
Collapse
Affiliation(s)
- Zinger Yang Loureiro
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shannon Joyce
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Javier Solivan-Rivera
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Anand Desai
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Pantos Skritakis
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Qin Yang
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Rachel Ziegler
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Denise Zhong
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Tammy T Nguyen
- Division of Vascular Surgery, Department of Surgery, UMass Memorial Medical Center, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
25
|
Ye J, Gao C, Liang Y, Hou Z, Shi Y, Wang Y. Characteristic and fate determination of adipose precursors during adipose tissue remodeling. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:13. [PMID: 37138165 PMCID: PMC10156890 DOI: 10.1186/s13619-023-00157-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/30/2022] [Indexed: 05/05/2023]
Abstract
Adipose tissues are essential for actively regulating systemic energy balance, glucose homeostasis, immune responses, reproduction, and longevity. Adipocytes maintain dynamic metabolic needs and possess heterogeneity in energy storage and supply. Overexpansion of adipose tissue, especially the visceral type, is a high risk for diabetes and other metabolic diseases. Changes in adipocytes, hypertrophy or hyperplasia, contribute to the remodeling of obese adipose tissues, accompanied by abundant immune cell accumulation, decreased angiogenesis, and aberrant extracellular matrix deposition. The process and mechanism of adipogenesis are well known, however, adipose precursors and their fate decision are only being defined with recent information available to decipher how adipose tissues generate, maintain, and remodel. Here, we discuss the key findings that identify adipose precursors phenotypically, with special emphasis on the intrinsic and extrinsic signals in instructing and regulating the fate of adipose precursors under pathophysiological conditions. We hope that the information in this review lead to novel therapeutic strategies to combat obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Jiayin Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Cheng Gao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yong Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
26
|
Ahmad B, Friar EP, Vohra MS, Khan N, Serpell CJ, Garrett MD, Loo JSE, Fong IL, Wong EH. Hydroxylated polymethoxyflavones reduce the activity of pancreatic lipase, inhibit adipogenesis and enhance lipolysis in 3T3-L1 mouse embryonic fibroblast cells. Chem Biol Interact 2023; 379:110503. [PMID: 37084996 DOI: 10.1016/j.cbi.2023.110503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
Hydroxylated polymethoxyflavones (HPMFs) have been shown to possess various anti-disease effects, including against obesity. This study investigates the anti-obesity effects of HPMFs in further detail, aiming to gain understanding of their mechanism of action in this context. The current study demonstrates that two HPMFs; 3'-hydroxy-5,7,4',5'-tetramethoxyflavone (3'OH-TetMF) and 4'-hydroxy-5,7,3',5'-tetramethoxyflavone (4'OH-TetMF) possess anti-obesity effects. They both significantly reduced pancreatic lipase activity in a competitive manner as demonstrated by molecular docking and kinetic studies. In cell studies, it was revealed that both of the HPMFs suppress differentiation of 3T3-L1 mouse embryonic fibroblast cells during the early stages of adipogenesis. They also reduced expression of key adipogenic and lipogenic marker genes, namely peroxisome proliferator-activated receptor-gamma (PPAR-γ), CCAAT/enhancer-binding protein α and β (C/EBP α and β), adipocyte binding protein 2 (aP2), fatty acid synthase (FASN), and sterol regulatory element-binding protein 1 (SREBF 1). They also enhanced the expression of cell cycle genes, i.e., cyclin D1 (CCND1) and C-Myc, and reduced cyclin A2 expression. When further investigated, it was also observed that these HPMFs accelerate lipid breakdown (lipolysis) and enhance lipolytic gene expression. Moreover, they also reduced the secretion of proteins (adipokines), including pro-inflammatory cytokines, from mature adipocytes. Taken together, this study concludes that these HPMFs have anti-obesity effects, which are worthy of further investigation.
Collapse
Affiliation(s)
- Bilal Ahmad
- School of Biosciences, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia
| | - Emily P Friar
- School of Chemistry and Forensic Science, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia
| | - Nasar Khan
- R3 Medical Research, 10045 East Dynamite Boulevard Suite 260, Scottsdale, AZ, 85262, United States
| | - Christopher J Serpell
- School of Chemistry and Forensic Science, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom.
| | - Michelle D Garrett
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, United Kingdom
| | - Jason Siau Ee Loo
- School of Pharmacy, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia
| | - Isabel Lim Fong
- Department of Paraclinical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak (UNIMAS), 94300, Kota Samarahan, Sarawak, Malaysia
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia.
| |
Collapse
|
27
|
Liu Q, Li C, Deng B, Gao P, Wang L, Li Y, Shiri M, Alkaifi F, Zhao J, Stephens JM, Simintiras CA, Francis J, Sun J, Fu X. Tcf21 marks visceral adipose mesenchymal progenitors and functions as a rate-limiting factor during visceral adipose tissue development. Cell Rep 2023; 42:112166. [PMID: 36857185 PMCID: PMC10208561 DOI: 10.1016/j.celrep.2023.112166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 01/01/2023] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Distinct locations of different white adipose depots suggest anatomy-specific developmental regulation, a relatively understudied concept. Here, we report a population of Tcf21 lineage cells (Tcf21 LCs) present exclusively in visceral adipose tissue (VAT) that dynamically contributes to VAT development and expansion. During development, the Tcf21 lineage gives rise to adipocytes. In adult mice, Tcf21 LCs transform into a fibrotic or quiescent state. Multiomics analyses show consistent gene expression and chromatin accessibility changes in Tcf21 LC, based on which we constructed a gene-regulatory network governing Tcf21 LC activities. Furthermore, single-cell RNA sequencing (scRNA-seq) identifies the heterogeneity of Tcf21 LCs. Loss of Tcf21 promotes the adipogenesis and developmental progress of Tcf21 LCs, leading to improved metabolic health in the context of diet-induced obesity. Mechanistic studies show that the inhibitory effect of Tcf21 on adipogenesis is at least partially mediated via Dlk1 expression accentuation.
Collapse
Affiliation(s)
- Qianglin Liu
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Chaoyang Li
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Buhao Deng
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA; Department of Animal Sciences, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Peidong Gao
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Leshan Wang
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Yuxia Li
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA
| | - Mohammad Shiri
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA
| | - Fozi Alkaifi
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA
| | - Junxing Zhao
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA; Department of Animal Sciences, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Jacqueline M Stephens
- Pennington Biomedical Research Center, Baton Rouge, LA, USA; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | | | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Jiangwen Sun
- Department of Computer Science, Old Dominion University, Norfolk, VA, USA.
| | - Xing Fu
- School of Animal Sciences, AgCenter, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
28
|
An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat Commun 2023; 14:1438. [PMID: 36922516 PMCID: PMC10017705 DOI: 10.1038/s41467-023-36983-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
To date, single-cell studies of human white adipose tissue (WAT) have been based on small cohort sizes and no cellular consensus nomenclature exists. Herein, we performed a comprehensive meta-analysis of publicly available and newly generated single-cell, single-nucleus, and spatial transcriptomic results from human subcutaneous, omental, and perivascular WAT. Our high-resolution map is built on data from ten studies and allowed us to robustly identify >60 subpopulations of adipocytes, fibroblast and adipogenic progenitors, vascular, and immune cells. Using these results, we deconvolved spatial and bulk transcriptomic data from nine additional cohorts to provide spatial and clinical dimensions to the map. This identified cell-cell interactions as well as relationships between specific cell subtypes and insulin resistance, dyslipidemia, adipocyte volume, and lipolysis upon long-term weight changes. Altogether, our meta-map provides a rich resource defining the cellular and microarchitectural landscape of human WAT and describes the associations between specific cell types and metabolic states.
Collapse
|
29
|
He Y, Wu N. Alternative Polyadenylation Results in mRNA Transcript Instability in Gestational Diabetes Mellitus. Diabetes Metab Syndr Obes 2023; 16:619-628. [PMID: 36915397 PMCID: PMC10008025 DOI: 10.2147/dmso.s400283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
OBJECTIVE To study the characteristics of selective polyadenylation (APA) in gestational diabetes mellitus (GDM) by poly(A) site sequencing and to explore the role of APA process in the pathogenesis of GDM. METHODS Three pregnant women diagnosed as GDM in our hospital were randomly selected as the GDM group, and three healthy pregnant women at the same time as the control group. The placental tissues of two groups of pregnant women after delivery were collected for high-throughput transcriptome sequencing (RNA-seq) and poly(A) site sequencing (PAS-seq) to screen differentially expressed genes and variable 3'UTR genes in GDM. Gene Ontology (GO) analysis and pathway analysis were used to analyze the functional classification and pathway of differential genes, and preliminarily explore the susceptible genes in GDM. RESULTS Compared with the control group, there were 202 TTS loci in the GDM group, including 103 genes with shortened TTS loci and 99 genes with delayed TTS loci. There were 57 genes with significant difference in TTS (P<0.05). Subsequently, we found that VCPIP1 and LGR4 were differentially expressed in RNA-seq. The genes in advance of TTS locus were enriched in biological processes such as cell development, protein transport and phosphorylation, signal transduction, etc. Delayed TTS genes are enriched in biological processes such as transcriptional regulation, cell migration and cycle, DNA repair and damage. CONCLUSION The abnormality of APA process may be involved in the occurrence and development of GDM. The genes with significantly different changes in TTS locus may become biomarkers or predictors for GDM to assess the incidence, disease progression and disease severity, and may also become potential targets for GDM treatment.
Collapse
Affiliation(s)
- Yujing He
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Na Wu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
- Department of Medical Service Quality, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
30
|
Maniyadath B, Zhang Q, Gupta RK, Mandrup S. Adipose tissue at single-cell resolution. Cell Metab 2023; 35:386-413. [PMID: 36889280 PMCID: PMC10027403 DOI: 10.1016/j.cmet.2023.02.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 03/09/2023]
Abstract
Adipose tissue exhibits remarkable plasticity with capacity to change in size and cellular composition under physiological and pathophysiological conditions. The emergence of single-cell transcriptomics has rapidly transformed our understanding of the diverse array of cell types and cell states residing in adipose tissues and has provided insight into how transcriptional changes in individual cell types contribute to tissue plasticity. Here, we present a comprehensive overview of the cellular atlas of adipose tissues focusing on the biological insight gained from single-cell and single-nuclei transcriptomics of murine and human adipose tissues. We also offer our perspective on the exciting opportunities for mapping cellular transitions and crosstalk, which have been made possible by single-cell technologies.
Collapse
Affiliation(s)
- Babukrishna Maniyadath
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Qianbin Zhang
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rana K Gupta
- Department of Internal Medicine, Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
31
|
A Wrong Fate Decision in Adipose Stem Cells upon Obesity. Cells 2023; 12:cells12040662. [PMID: 36831329 PMCID: PMC9954614 DOI: 10.3390/cells12040662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Progress has been made in identifying stem cell aging as a pathological manifestation of a variety of diseases, including obesity. Adipose stem cells (ASCs) play a core role in adipocyte turnover, which maintains tissue homeostasis. Given aberrant lineage determination as a feature of stem cell aging, failure in adipogenesis is a culprit of adipose hypertrophy, resulting in adiposopathy and related complications. In this review, we elucidate how ASC fails in entering adipogenic lineage, with a specific focus on extracellular signaling pathways, epigenetic drift, metabolic reprogramming, and mechanical stretch. Nonetheless, such detrimental alternations can be reversed by guiding ASCs towards adipogenesis. Considering the pathological role of ASC aging in obesity, targeting adipogenesis as an anti-obesity treatment will be a key area of future research, and a strategy to rejuvenate tissue stem cell will be capable of alleviating metabolic syndrome.
Collapse
|
32
|
MME + fibro-adipogenic progenitors are the dominant adipogenic population during fatty infiltration in human skeletal muscle. Commun Biol 2023; 6:111. [PMID: 36707617 PMCID: PMC9883500 DOI: 10.1038/s42003-023-04504-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/18/2023] [Indexed: 01/29/2023] Open
Abstract
Fatty infiltration, the ectopic deposition of adipose tissue within skeletal muscle, is mediated via the adipogenic differentiation of fibro-adipogenic progenitors (FAPs). We used single-nuclei and single-cell RNA sequencing to characterize FAP heterogeneity in patients with fatty infiltration. We identified an MME+ FAP subpopulation which, based on ex vivo characterization as well as transplantation experiments, exhibits high adipogenic potential. MME+ FAPs are characterized by low activity of WNT, known to control adipogenic commitment, and are refractory to the inhibitory role of WNT activators. Using preclinical models for muscle damage versus fatty infiltration, we show that many MME+ FAPs undergo apoptosis during muscle regeneration and differentiate into adipocytes under pathological conditions, leading to a reduction in their abundance. Finally, we utilized the varying fat infiltration levels in human hip muscles and found less MME+ FAPs in fatty infiltrated human muscle. Altogether, we have identified the dominant adipogenic FAP subpopulation in skeletal muscle.
Collapse
|
33
|
Chao Y, Gao L, Wang X, Cai Y, Shu Y, Zou X, Qin Y, Hu C, Dai Y, Zhu M, Shen Z, Zou C. Dysregulated adipose tissue expansion and impaired adipogenesis in Prader-Willi syndrome children before obesity-onset. Metabolism 2022; 136:155295. [PMID: 36007622 DOI: 10.1016/j.metabol.2022.155295] [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: 04/30/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Prader-Willi syndrome (PWS) is a rare genetic imprinting disorder resulting from the expression loss of genes on the paternally inherited chromosome 15q11-13. Early-onset life-thriving obesity and hyperphagia represent the clinical hallmarks of PWS. The noncoding RNA gene SNORD116 within the minimal PWS genetic lesion plays a critical role in the pathogenesis of the syndrome. Despite advancements in understanding the genetic basis for PWS, the pathophysiology of obesity development in PWS remains largely uncharacterized. Here, we aimed to investigate the signatures of adipose tissue development and expansion pathways and associated adipose biology in PWS children without obesity-onset at an early stage, mainly from the perspective of the adipogenesis process, and further elucidate the underlying molecular mechanisms. METHODS We collected inguinal (subcutaneous) white adipose tissues (ingWATs) from phase 1 PWS and healthy children with normal weight aged from 6 M to 2 Y. Adipose morphology and histological characteristics were assessed. Primary adipose stromal vascular fractions (SVFs) were isolated, cultured in vitro, and used to determine the capacity and function of white and beige adipogenic differentiation. High-throughput RNA-sequencing (RNA-seq) was performed in adipose-derived mesenchymal stem cells (AdMSCs) to analyze transcriptome signatures in PWS subjects. Transient repression of SNORD116 was conducted to evaluate its functional relevance in adipogenesis. The changes in alternative pre-mRNA splicing were investigated in PWS and SNORD116 deficient cells. RESULTS In phase 1 PWS children, impaired white adipose tissue (WAT) development and unusual fat expansion occurred long before obesity onset, which was characterized by the massive enlargement of adipocytes accompanied by increased apoptosis. White and beige adipogenesis programs were impaired and differentiated adipocyte functions were disturbed in PWS-derived SVFs, despite increased proliferation capacity, which were consistent with the results of RNA-seq analysis of PWS AdMSCs. We also experimentally validated disrupted beige adipogenesis in adipocytes with transient SNORD116 downregulation. The transcript and protein levels of PPARγ, the adipogenesis master regulator, were significantly lower in PWS than in control AdMSCs as well as in SNORD116 deficient AdMSCs/adipocytes than in scramble (Scr) cells, resulting in the inhibited adipogenic program. Additionally, through RNA-seq, we observed aberrant transcriptome-wide alterations in alternative RNA splicing patterns in PWS cells mediated by SNORD116 loss and specifically identified a changed PRDM16 gene splicing profile in vitro. CONCLUSIONS Imbalance in the WAT expansion pathway and developmental disruption are primary defects in PWS displaying aberrant adipocyte hypertrophy and impaired adipogenesis process, in which SNORD116 deficiency plays a part. Our findings suggest that dysregulated adiposity specificity existing at an early phase is a potential pathological mechanism exacerbating hyperphagic obesity onset in PWS. This mechanistic evidence on adipose biology in young PWS patients expands knowledge regarding the pathogenesis of PWS obesity and may aid in developing a new therapeutic strategy targeting disturbed adipogenesis and driving AT plasticity to combat abnormal adiposity and associated metabolic disorders for PWS patients.
Collapse
Affiliation(s)
- Yunqi Chao
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Lei Gao
- Department of Urology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Xiangzhi Wang
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Yuqing Cai
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Yingying Shu
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Xinyi Zou
- Zhejiang University City College, Hangzhou 310015, Zhejiang, China
| | - Yifang Qin
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Chenxi Hu
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Yangli Dai
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Mingqiang Zhu
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Zheng Shen
- Lab Center, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China
| | - Chaochun Zou
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, Zhejiang, China.
| |
Collapse
|
34
|
Yang J, Vamvini M, Nigro P, Ho LL, Galani K, Alvarez M, Tanigawa Y, Renfro A, Carbone NP, Laakso M, Agudelo LZ, Pajukanta P, Hirshman MF, Middelbeek RJW, Grove K, Goodyear LJ, Kellis M. Single-cell dissection of the obesity-exercise axis in adipose-muscle tissues implies a critical role for mesenchymal stem cells. Cell Metab 2022; 34:1578-1593.e6. [PMID: 36198295 PMCID: PMC9558082 DOI: 10.1016/j.cmet.2022.09.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/29/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022]
Abstract
Exercise training is critical for the prevention and treatment of obesity, but its underlying mechanisms remain incompletely understood given the challenge of profiling heterogeneous effects across multiple tissues and cell types. Here, we address this challenge and opposing effects of exercise and high-fat diet (HFD)-induced obesity at single-cell resolution in subcutaneous and visceral white adipose tissue and skeletal muscle in mice with diet and exercise training interventions. We identify a prominent role of mesenchymal stem cells (MSCs) in obesity and exercise-induced tissue adaptation. Among the pathways regulated by exercise and HFD in MSCs across the three tissues, extracellular matrix remodeling and circadian rhythm are the most prominent. Inferred cell-cell interactions implicate within- and multi-tissue crosstalk centered around MSCs. Overall, our work reveals the intricacies and diversity of multi-tissue molecular responses to exercise and obesity and uncovers a previously underappreciated role of MSCs in tissue-specific and multi-tissue beneficial effects of exercise.
Collapse
Affiliation(s)
- Jiekun Yang
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Maria Vamvini
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Pasquale Nigro
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Li-Lun Ho
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kyriakitsa Galani
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yosuke Tanigawa
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashley Renfro
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nicholas P Carbone
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Leandro Z Agudelo
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Roeland J W Middelbeek
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Kevin Grove
- Novo Nordisk Research Center, Seattle, WA, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
35
|
Yang L, Yue W, Zhang H, Gao Y, Yang L, Li L. The role of roof plate-specific spondins in liver homeostasis and disease. LIVER RESEARCH 2022; 6:139-145. [PMID: 39958194 PMCID: PMC11791806 DOI: 10.1016/j.livres.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/05/2022] [Accepted: 09/02/2022] [Indexed: 02/16/2023]
Abstract
As evolutionarily conserved signals, roof plate-specific spondins (R-spondins; RSPOs) are a family with four members (RSPO1-4) exerting distinctly different functions. RSPOs have five receptors and correlate with different signaling pathways through these receptors and then perform various functions. Moreover, their best-known molecular function is the capacity to enhance WNT signaling pathways, which play critical roles in several processes. A recent study shows that RSPOs not only potentiate the WNT/beta (β)-catenin signaling pathway but are also involved in the WNT/planar cell polarity signaling pathway. RSPOs influence liver homeostasis and the development of multiple liver diseases. RSPO1 increases cell proliferation, protects hepatocytes from injury, improves liver regenerative potential, and affects liver metabolic zonation. RSPO2 not only regulates proliferation-associated genes and promotes differentiation in the liver but also participates in liver fibrosis through the WNT/β-catenin signaling pathway. RSPO3 is a key determinant of proper liver function, such as promoting hepatocyte regeneration and maintaining liver zonation. RSPO3 is upregulated in liver fibrosis and livers of patients with non-alcoholic steatohepatitis. Besides, RSPO2 and RSPO3 are confirmed as oncogenes and involved in the occurrence of liver cancer. The role of RSPO4 in the liver remains unclear. In this review, the structural and biochemical properties of RSPOs and their receptors and their roles in liver homeostasis and disease are summarized.
Collapse
Affiliation(s)
- Le Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Wenhui Yue
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Hang Zhang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Yue Gao
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| |
Collapse
|
36
|
Zachara M, Rainer PY, Hashimi H, Russeil JM, Alpern D, Ferrero R, Litovchenko M, Deplancke B. Mammalian adipogenesis regulator (Areg) cells use retinoic acid signalling to be non- and anti-adipogenic in age-dependent manner. EMBO J 2022; 41:e108206. [PMID: 35996853 PMCID: PMC9475530 DOI: 10.15252/embj.2021108206] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/09/2022] Open
Abstract
Adipose stem and precursor cells (ASPCs) give rise to adipocytes and determine the composition and plasticity of adipose tissue. Recently, several studies have demonstrated that ASPCs partition into at least three distinct cell subpopulations, including the enigmatic CD142+ cells. An outstanding challenge is to functionally characterise this population, as discrepant properties, from adipogenic to non- and anti-adipogenic, have been reported for these cells. To resolve these phenotypic ambiguities, we characterised mammalian subcutaneous CD142+ ASPCs across various experimental conditions, demonstrating that CD142+ ASPCs exhibit high molecular and phenotypic robustness. Specifically, we find these cells to be firmly non- and anti-adipogenic both in vitro and in vivo, with their inhibitory signals also impacting adipogenic human cells. However, these CD142+ ASPC-specific properties exhibit surprising temporal phenotypic alterations, and emerge only in an age-dependent manner. Finally, using multi-omic and functional assays, we show that the inhibitory nature of these adipogenesis-regulatory CD142+ ASPCs (Aregs) is driven by specifically expressed secretory factors that cooperate with the retinoic acid signalling pathway to transform the adipogenic state of CD142- ASPCs into a non-adipogenic, Areg-like state.
Collapse
Affiliation(s)
- Magda Zachara
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Pernille Y Rainer
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Horia Hashimi
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Julie M Russeil
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Radiana Ferrero
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
37
|
Lecoutre S, Lambert M, Drygalski K, Dugail I, Maqdasy S, Hautefeuille M, Clément K. Importance of the Microenvironment and Mechanosensing in Adipose Tissue Biology. Cells 2022; 11:cells11152310. [PMID: 35954152 PMCID: PMC9367348 DOI: 10.3390/cells11152310] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
The expansion of adipose tissue is an adaptive mechanism that increases nutrient buffering capacity in response to an overall positive energy balance. Over the course of expansion, the adipose microenvironment undergoes continual remodeling to maintain its structural and functional integrity. However, in the long run, adipose tissue remodeling, typically characterized by adipocyte hypertrophy, immune cells infiltration, fibrosis and changes in vascular architecture, generates mechanical stress on adipose cells. This mechanical stimulus is then transduced into a biochemical signal that alters adipose function through mechanotransduction. In this review, we describe the physical changes occurring during adipose tissue remodeling, and how they regulate adipose cell physiology and promote obesity-associated dysfunction in adipose tissue.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Mélanie Lambert
- Labex Inflamex, Université Sorbonne Paris Nord, INSERM, F-93000 Bobigny, France;
| | - Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet Hospital, C2-94, 14186 Stockholm, Sweden;
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR 7622), IBPS, Sorbonne Université, F-75005 Paris, France;
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, F-75013 Paris, France
- Correspondence: or
| |
Collapse
|
38
|
Burl RB, Rondini EA, Wei H, Pique-Regi R, Granneman JG. Deconstructing cold-induced brown adipocyte neogenesis in mice. eLife 2022; 11:e80167. [PMID: 35848799 PMCID: PMC9348851 DOI: 10.7554/elife.80167] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/15/2022] [Indexed: 11/21/2022] Open
Abstract
Cold exposure triggers neogenesis in classic interscapular brown adipose tissue (iBAT) that involves activation of β1-adrenergic receptors, proliferation of PDGFRA+ adipose tissue stromal cells (ASCs), and recruitment of immune cells whose phenotypes are presently unknown. Single-cell RNA-sequencing (scRNA-seq) in mice identified three ASC subpopulations that occupied distinct tissue locations. Of these, interstitial ASC1 were found to be direct precursors of new brown adipocytes (BAs). Surprisingly, knockout of β1-adrenergic receptors in ASCs did not prevent cold-induced neogenesis, whereas pharmacological activation of the β3-adrenergic receptor on BAs was sufficient, suggesting that signals derived from mature BAs indirectly trigger ASC proliferation and differentiation. In this regard, cold exposure induced the delayed appearance of multiple macrophage and dendritic cell populations whose recruitment strongly correlated with the onset and magnitude of neogenesis across diverse experimental conditions. High-resolution immunofluorescence and single-molecule fluorescence in situ hybridization demonstrated that cold-induced neogenesis involves dynamic interactions between ASC1 and recruited immune cells that occur on the micrometer scale in distinct tissue regions. Our results indicate that neogenesis is not a reflexive response of progenitors to β-adrenergic signaling, but rather is a complex adaptive response to elevated metabolic demand within brown adipocytes.
Collapse
Affiliation(s)
- Rayanne B Burl
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Elizabeth Ann Rondini
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
- Center for Integrative Metabolic and Endocrine Research, Wayne State UniversityDetroitUnited States
| | - Hongguang Wei
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
- Center for Integrative Metabolic and Endocrine Research, Wayne State UniversityDetroitUnited States
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
- Center for Integrative Metabolic and Endocrine Research, Wayne State UniversityDetroitUnited States
| |
Collapse
|
39
|
Franco CN, Noe MM, Albrecht LV. Metabolism and Endocrine Disorders: What Wnt Wrong? Front Endocrinol (Lausanne) 2022; 13:887037. [PMID: 35600583 PMCID: PMC9120667 DOI: 10.3389/fendo.2022.887037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Abstract
A fundamental question in cell biology underlies how nutrients are regenerated to maintain and renew tissues. Physiologically, the canonical Wnt signaling is a vital pathway for cell growth, tissue remodeling, and organ formation; pathologically, Wnt signaling contributes to the development of myriad human diseases such as cancer. Despite being the focus of intense research, how Wnt intersects with the metabolic networks to promote tissue growth and remodeling has remained mysterious. Our understanding of metabolism has been revolutionized by technological advances in the fields of chemical biology, metabolomics, and live microscopy that have now made it possible to visualize and manipulate metabolism in living cells and tissues. The application of these toolsets to innovative model systems have propelled the Wnt field into new realms at the forefront answering the most pressing paradigms of cell metabolism in health and disease states. Elucidating the basis of Wnt signaling and metabolism in a cell-type and tissue-specific manner will provide a powerful base of knowledge for both basic biomedical fields and clinician scientists, and has the promise to generate new, transformative therapies in disease and even processes of aging.
Collapse
Affiliation(s)
- Carolina N. Franco
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA, United States
| | - May M. Noe
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA, United States
| | - Lauren V. Albrecht
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA, United States
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
40
|
Duerre DJ, Galmozzi A. Deconstructing Adipose Tissue Heterogeneity One Cell at a Time. Front Endocrinol (Lausanne) 2022; 13:847291. [PMID: 35399946 PMCID: PMC8990929 DOI: 10.3389/fendo.2022.847291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/28/2022] [Indexed: 12/26/2022] Open
Abstract
As a central coordinator of physiologic metabolism, adipose tissue has long been appreciated as a highly plastic organ that dynamically responds to environmental cues. Once thought of as a homogenous storage depot, recent advances have enabled deep characterizations of the underlying structure and composition of adipose tissue depots. As the obesity and metabolic disease epidemics continue to accelerate due to modern lifestyles and an aging population, elucidation of the underlying mechanisms that control adipose and systemic homeostasis are of critical importance. Within the past decade, the emergence of deep cell profiling at tissue- and, recently, single-cell level has furthered our understanding of the complex dynamics that contribute to tissue function and their implications in disease development. Although many paradigm-shifting findings may lie ahead, profound advances have been made to forward our understanding of the adipose tissue niche in both health and disease. Now widely accepted as a highly heterogenous organ with major roles in metabolic homeostasis, endocrine signaling, and immune function, the study of adipose tissue dynamics has reached a new frontier. In this review, we will provide a synthesis of the latest advances in adipose tissue biology made possible by the use of single-cell technologies, the impact of epigenetic mechanisms on adipose function, and suggest what next steps will further our understanding of the role that adipose tissue plays in systemic physiology.
Collapse
Affiliation(s)
- Dylan J. Duerre
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Andrea Galmozzi
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
41
|
Cannavino J, Gupta RK. Cellular and molecular brakes on adipogenesis. Nat Metab 2022; 4:13-14. [PMID: 35027769 DOI: 10.1038/s42255-021-00519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jessica Cannavino
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
42
|
Filipowska J, Kondegowda NG, Leon-Rivera N, Dhawan S, Vasavada RC. LGR4, a G Protein-Coupled Receptor With a Systemic Role: From Development to Metabolic Regulation. Front Endocrinol (Lausanne) 2022; 13:867001. [PMID: 35707461 PMCID: PMC9190282 DOI: 10.3389/fendo.2022.867001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
Leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4/GPR48), a member of the GPCR (G protein-coupled receptors) superfamily, subfamily B, is a common intestinal crypt stem cell marker. It binds R-spondins/Norrin as classical ligands and plays a crucial role in Wnt signaling potentiation. Interaction between LGR4 and R-spondins initiates many Wnt-driven developmental processes, e.g., kidney, eye, or reproductive tract formation, as well as intestinal crypt (Paneth) stem cell pool maintenance. Besides the well-described role of LGR4 in development, several novel functions of this receptor have recently been discovered. In this context, LGR4 was indicated to participate in TGFβ and NFκB signaling regulation in hematopoietic precursors and intestinal cells, respectively, and found to be a new, alternative receptor for RANKL (Receptor Activator of NF kappa B Ligand) in bone cells. LGR4 inhibits the process of osteoclast differentiation, by antagonizing the interaction between RANK (Receptor Activator of NF kappa B) and its ligand-RANKL. It is also known to trigger anti-inflammatory responses in different tissues (liver, intestine, cardiac cells, and skin), serve as a sensor of the circadian clock in the liver, regulate adipogenesis and energy expenditure in adipose tissue and skeletal muscles, respectively. The extracellular domain of LGR4 (LGR4-ECD) has emerged as a potential new therapeutic for osteoporosis and cancer. LGR4 integrates different signaling pathways and regulates various cellular processes vital for maintaining whole-body homeostasis. Yet, the role of LGR4 in many cell types (e.g. pancreatic beta cells) and diseases (e.g., diabetes) remains to be elucidated. Considering the broad spectrum of LGR4 actions, this review aims to discuss both canonical and novel roles of LGR4, with emphasis on emerging research directions focused on this receptor.
Collapse
|