1
|
Chen Q, Zheng J, Bian Q. Cell Fate Regulation During the Development of Infantile Hemangioma. J Invest Dermatol 2025; 145:266-279. [PMID: 39023471 DOI: 10.1016/j.jid.2024.06.1275] [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: 02/29/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 07/20/2024]
Abstract
As the most common benign vascular tumor in infants, infantile hemangioma (IH) is characterized by rapid growth and vasculogenesis early in infancy, followed by spontaneous involution into fibrofatty tissues over time. Extensive evidence suggests that IH originates from hemangioma stem cells (HemSCs), a group of stem cells with clonal expansion and multi-directional differentiation capacity. However, the intricate mechanisms governing the cell fate transition of HemSCs during IH development remain elusive. Here we comprehensively examine the cellular composition of IH, emphasizing the nuanced properties of various IH cell types and their correlation with the clinical features of the tumor. We also summarize the current understanding of the regulatory pathways directing HemSC differentiation into endothelial cells (ECs), pericytes, and adipocytes throughout the stages of IH progression and involution. Furthermore, we discuss recent advances in unraveling the transcriptional and epigenetic regulation of EC and adipocyte development under physiological conditions, which offer crucial perspectives for understanding IH pathogenesis.
Collapse
Affiliation(s)
- Qiming Chen
- Department of Oromaxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jiawei Zheng
- Department of Oromaxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China.
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Ma X, Zhang D, Yang Z, Sun M, Gao N, Mei C, Zan L. bta-miR-484 Inhibits Bovine Intramuscular Adipogenesis by Regulating Mitotic Clonal Expansion via the MAP3K9/JNK/CCND1 Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:1062-1074. [PMID: 39719059 DOI: 10.1021/acs.jafc.4c07956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Intramuscular fat (IMF) content is a critical indicator of the beef nutritional value and flavor. In this study, we focused on bta-miR-484, a microRNA that is differentially expressed during the adipogenic differentiation of bovine intramuscular adipocytes and is negatively correlated with the IMF content across different cattle breeds. Our findings demonstrate that bta-miR-484 inhibits adipogenic differentiation without altering the fatty acid composition of bovine intramuscular adipocytes. miRNA pull-down and dual-luciferase reporter assays confirmed that MAP3K9 is a target gene of bta-miR-484. Furthermore, bta-miR-484 suppresses the JNK signaling pathway by targeting MAP3K9, leading to decreased CCND1 expression, which impedes the mitotic clonal expansion (MCE) process and inhibits intramuscular adipocyte differentiation. In summary, this study uncovers a novel mechanism by which bta-miR-484 regulates bovine IMF content and provides the first exploration of MCE during intramuscular adipocyte adipogenic differentiation. These findings offer valuable theoretical insights into beef cattle breeding and molecular improvements.
Collapse
Affiliation(s)
- Xinhao Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Dianqi Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhimei Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Meijun Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Ni Gao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Chugang Mei
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| |
Collapse
|
3
|
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 2025:10.1038/s41574-024-01071-y. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
4
|
Lambert J, Kovilakath A, Jamil M, Valentine Y, Anderson A, Montefusco D, Cowart LA. Sphingosine kinase 1 is induced by glucocorticoids in adipose derived stem cells and enhances glucocorticoid mediated signaling in adipose expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612482. [PMID: 39314417 PMCID: PMC11419133 DOI: 10.1101/2024.09.13.612482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Sphingosine kinase 1 (SphK1) plays a crucial role in regulating metabolic pathways within adipocytes and is elevated in the adipose tissue of obese mice. While previous studies have reported both pro- and inhibitory effects of SphK1 and its product, sphingosine-1-phosphate (S1P), on adipogenesis, the precise mechanisms remain unclear. This study explores the timing and downstream effects of SphK1/S1P expression and activation during in vitro adipogenesis. We demonstrate that the synthetic glucocorticoid dexamethasone robustly induces SphK1 expression, suggesting its involvement in glucocorticoid-dependent signaling during adipogenesis. Notably, the activation of C/EBPδ, a key gene in early adipogenesis and a target of glucocorticoids, is diminished in SphK1-/- adipose-derived stem cells (ADSCs). Furthermore, glucocorticoid administration promotes adipose tissue expansion via SphK1 in a depot-specific manner. Although adipose expansion still occurs in SphK1-/- mice, it is significantly reduced. These findings indicate that while SphK1 is not essential for adipogenesis, it enhances early gene activation, thereby facilitating adipose tissue expansion.
Collapse
Affiliation(s)
- Johana Lambert
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Departments of Biochemistry and Molecular Biology Medical University of South Carolina, Charleston, SC, USA
| | - Anna Kovilakath
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Maryam Jamil
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Yolander Valentine
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrea Anderson
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Departments of Biochemistry and Molecular Biology Medical University of South Carolina, Charleston, SC, USA
| | - David Montefusco
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, USA
| |
Collapse
|
5
|
Anaya ES, de Groot EL, Lydon JP, Pangas SA, Hartig SM. Contributions of white adipose tissue to energy requirements for female reproduction. Trends Endocrinol Metab 2024; 35:809-820. [PMID: 38749883 PMCID: PMC11387141 DOI: 10.1016/j.tem.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 09/12/2024]
Abstract
Body composition impacts female fertility and there are established relationships between adipose tissue and the reproductive system. Maintaining functional adipose tissue is vital for meeting the energetic demands during the reproductive process, from ovulation to delivery and lactation. White adipose tissue (WAT) shows plastic responses to daily physiology and secretes diverse adipokines that affect the hypothalamic-pituitary-ovarian axis, but many other interorgan interactions remain to be determined. This review summarizes the current state of research on the dialogue between WAT and the female reproductive system, focusing on the impact of this crosstalk on ovarian and endometrial factors essential for fecundity.
Collapse
Affiliation(s)
- Elizabeth S Anaya
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Evelyn L de Groot
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Stephanie A Pangas
- Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Sean M Hartig
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
6
|
Dowker-Key PD, Jadi PK, Gill NB, Hubbard KN, Elshaarrawi A, Alfatlawy ND, Bettaieb A. A Closer Look into White Adipose Tissue Biology and the Molecular Regulation of Stem Cell Commitment and Differentiation. Genes (Basel) 2024; 15:1017. [PMID: 39202377 PMCID: PMC11353785 DOI: 10.3390/genes15081017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
White adipose tissue (WAT) makes up about 20-25% of total body mass in healthy individuals and is crucial for regulating various metabolic processes, including energy metabolism, endocrine function, immunity, and reproduction. In adipose tissue research, "adipogenesis" is commonly used to refer to the process of adipocyte formation, spanning from stem cell commitment to the development of mature, functional adipocytes. Although, this term should encompass a wide range of processes beyond commitment and differentiation, to also include other stages of adipose tissue development such as hypertrophy, hyperplasia, angiogenesis, macrophage infiltration, polarization, etc.… collectively, referred to herein as the adipogenic cycle. The term "differentiation", conversely, should only be used to refer to the process by which committed stem cells progress through distinct phases of subsequent differentiation. Recognizing this distinction is essential for accurately interpreting research findings on the mechanisms and stages of adipose tissue development and function. In this review, we focus on the molecular regulation of white adipose tissue development, from commitment to terminal differentiation, and examine key functional aspects of WAT that are crucial for normal physiology and systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Presley D. Dowker-Key
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Praveen Kumar Jadi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Nicholas B. Gill
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Katelin N. Hubbard
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Elshaarrawi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Naba D. Alfatlawy
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN 37996-0840, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996-0840, USA
| |
Collapse
|
7
|
Yu S, Yu H, Wang J, Liu H, Guo J, Wang S, Mei C, Zan L. LEP inhibits intramuscular adipogenesis through the AMPK signaling pathway in vitro. FASEB J 2024; 38:e23836. [PMID: 39044640 DOI: 10.1096/fj.202400590rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
Leptin can indirectly regulate fatty-acid metabolism and synthesis in muscle in vivo and directly in incubated muscle ex vivo. In addition, non-synonymous mutations in the bovine leptin gene (LEP) are associated with carcass intramuscular fat (IMF) content. However, the effects of LEP on lipid synthesis of adipocytes have not been clearly studied at the cellular level. Therefore, this study focused on bovine primary intramuscular preadipocytes to investigate the effects of LEP on the proliferation and differentiation of intramuscular preadipocytes, as well as its regulatory mechanism in lipid synthesis. The results showed that both the LEP and leptin receptor gene (LEPR) were highly expressed in IMF tissues, and their mRNA expression levels were positively correlated at different developmental stages of intramuscular preadipocytes. The overexpression of LEP inhibited the proliferation and differentiation of intramuscular preadipocytes, while interference with LEP had the opposite effect. Additionally, LEP significantly promoted the phosphorylation level of AMPKα by promoting the protein expression of CAMKK2. Meanwhile, rescue experiments showed that the increasing effect of AMPK inhibitors on the number of intramuscular preadipocytes was significantly weakened by the overexpression of LEP. Furthermore, the overexpression of LEP could weaken the promoting effect of AMPK inhibitor on triglyceride content and droplet accumulation, and prevent the upregulation of adipogenic protein expression (SREBF1, FABP4, FASN, and ACCα) caused by AMPK inhibitor. Taken together, LEP acted on the AMPK signaling pathway by regulating the protein expression of CAMKK2, thereby downregulating the expression of proliferation-related and adipogenic-related genes and proteins, ultimately reducing intramuscular adipogenesis.
Collapse
Affiliation(s)
- Shengchen Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hengwei Yu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jianfang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Haibing Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Juntao Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Sihu Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chugang Mei
- College of Grassland Agriculture, Northwest A&F University, Yangling, China
- National Beef Cattle Improvement Center, Yangling, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
- National Beef Cattle Improvement Center, Yangling, China
| |
Collapse
|
8
|
Chen J, Pan Y, Lu Y, Fang X, Ma T, Chen X, Wang Y, Fang X, Zhang C, Song C. The Function and Mechanism of Long Noncoding RNAs in Adipogenic Differentiation. Genes (Basel) 2024; 15:875. [PMID: 39062654 PMCID: PMC11275360 DOI: 10.3390/genes15070875] [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: 05/21/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Adipocytes are crucial for maintaining energy balance. Adipocyte differentiation involves distinct stages, including the orientation stage, clone amplification stage, clone amplification termination stage, and terminal differentiation stage. Understanding the regulatory mechanisms governing adipogenic differentiation is essential for comprehending the physiological processes and identifying potential biomarkers and therapeutic targets for metabolic diseases, ultimately improving glucose and fat metabolism. Adipogenic differentiation is influenced not only by key factors such as hormones, the peroxisome proliferator-activated receptor (PPAR) family, and the CCATT enhancer-binding protein (C/EBP) family but also by noncoding RNA, including microRNA (miRNA), long noncoding RNA (lncRNA), and circular RNA (circRNA). Among these, lncRNA has been identified as a significant regulator in adipogenic differentiation. Research has demonstrated various ways in which lncRNAs contribute to the molecular mechanisms of adipogenic differentiation. Throughout the adipogenesis process, lncRNAs modulate adipocyte differentiation and development by influencing relevant signaling pathways and transcription factors. This review provides a brief overview of the function and mechanism of lncRNAs in adipogenic differentiation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Chunlei Zhang
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (J.C.); (Y.P.); (Y.L.); (X.F.); (T.M.); (X.C.); (Y.W.); (X.F.)
| | - Chengchuang Song
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; (J.C.); (Y.P.); (Y.L.); (X.F.); (T.M.); (X.C.); (Y.W.); (X.F.)
| |
Collapse
|
9
|
Qian S, Zhang C, Tang Y, Dai M, He Z, Ma H, Wang L, Yang Q, Liu Y, Xu W, Zhang Z, Tang QQ. A single-cell sequence analysis of mouse subcutaneous white adipose tissue reveals dynamic changes during weaning. Commun Biol 2024; 7:787. [PMID: 38951550 PMCID: PMC11217364 DOI: 10.1038/s42003-024-06448-3] [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: 05/10/2023] [Accepted: 06/12/2024] [Indexed: 07/03/2024] Open
Abstract
Adipose tissue development begins in the fetal period, and continues to expand after birth. Dysregulation of adipose tissue during weaning may predispose individuals to lifelong metabolic disorders. However, the developmental remodeling of adipose tissue during weaning remains largely unexplored. Here we comprehensively compare the changes in mouse subcutaneous white adipose tissue from 7 days after birth to 7 days after weaning using single-cell RNA sequencing along with other molecular and histologic assays. We characterize the developmental trajectory of preadipocytes and indicate the commitment of preadipocytes with beige potential during weaning. Meanwhile, we find immune cells unique to weaning period, whose expression of extracellular matrix proteins implies potential regulation on preadipocyte. Finally, the strongest cell-cell interaction during weaning determined by the TGFβ ligand-receptor pairs is between preadipocytes and endotheliocytes. Our results provide a detailed and unbiased cellular landscape and offer insights into the potential regulation of adipose tissue remodeling during weaning.
Collapse
Affiliation(s)
- Shuwen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chenyang Zhang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Mengyuan Dai
- Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhihui He
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hong Ma
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Linyuan Wang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qiqi Yang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Xu
- Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhao Zhang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Boychenko S, Egorova VS, Brovin A, Egorov AD. White-to-Beige and Back: Adipocyte Conversion and Transcriptional Reprogramming. Pharmaceuticals (Basel) 2024; 17:790. [PMID: 38931457 PMCID: PMC11206576 DOI: 10.3390/ph17060790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Obesity has become a pandemic, as currently more than half a billion people worldwide are obese. The etiology of obesity is multifactorial, and combines a contribution of hereditary and behavioral factors, such as nutritional inadequacy, along with the influences of environment and reduced physical activity. Two types of adipose tissue widely known are white and brown. While white adipose tissue functions predominantly as a key energy storage, brown adipose tissue has a greater mass of mitochondria and expresses the uncoupling protein 1 (UCP1) gene, which allows thermogenesis and rapid catabolism. Even though white and brown adipocytes are of different origin, activation of the brown adipocyte differentiation program in white adipose tissue cells forces them to transdifferentiate into "beige" adipocytes, characterized by thermogenesis and intensive lipolysis. Nowadays, researchers in the field of small molecule medicinal chemistry and gene therapy are making efforts to develop new drugs that effectively overcome insulin resistance and counteract obesity. Here, we discuss various aspects of white-to-beige conversion, adipose tissue catabolic re-activation, and non-shivering thermogenesis.
Collapse
Affiliation(s)
- Stanislav Boychenko
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| | - Vera S. Egorova
- Biotechnology Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia
| | - Andrew Brovin
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| | - Alexander D. Egorov
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| |
Collapse
|
11
|
Gao Z, Santos RB, Rupert J, Van Drunen R, Yu Y, Eckel‐Mahan K, Kolonin MG. Endothelial-specific telomerase inactivation causes telomere-independent cell senescence and multi-organ dysfunction characteristic of aging. Aging Cell 2024; 23:e14138. [PMID: 38475941 PMCID: PMC11296101 DOI: 10.1111/acel.14138] [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: 11/02/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
It has remained unclear how aging of endothelial cells (EC) contributes to pathophysiology of individual organs. Cell senescence results in part from inactivation of telomerase (TERT). Here, we analyzed mice with Tert knockout specifically in EC. Tert loss in EC induced transcriptional changes indicative of senescence and tissue hypoxia in EC and in other cells. We demonstrate that EC-Tert-KO mice have leaky blood vessels. The blood-brain barrier of EC-Tert-KO mice is compromised, and their cognitive function is impaired. EC-Tert-KO mice display reduced muscle endurance and decreased expression of enzymes responsible for oxidative metabolism. Our data indicate that Tert-KO EC have reduced mitochondrial content and function, which results in increased dependence on glycolysis. Consistent with this, EC-Tert-KO mice have metabolism changes indicative of increased glucose utilization. In EC-Tert-KO mice, expedited telomere attrition is observed for EC of adipose tissue (AT), while brain and skeletal muscle EC have normal telomere length but still display features of senescence. Our data indicate that the loss of Tert causes EC senescence in part through a telomere length-independent mechanism undermining mitochondrial function. We conclude that EC-Tert-KO mice is a model of expedited vascular senescence recapitulating the hallmarks aging, which can be useful for developing revitalization therapies.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rafael Bravo Santos
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Joseph Rupert
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rachel Van Drunen
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
12
|
Shi Z, Xiong S, Hu R, Wang Z, Park J, Qian Y, Wang J, Bhalla P, Velupally N, Song Q, Song Z, Jeon MS, Zhang KK, Xie L, Layden BT, Ong SG, Jiang Y. The Notch-PDGFRβ axis suppresses brown adipocyte progenitor differentiation in early post-natal mice. Dev Cell 2024; 59:1233-1251.e5. [PMID: 38569546 DOI: 10.1016/j.devcel.2024.03.012] [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: 05/23/2023] [Revised: 12/08/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively explored. Here, through in vivo lineage tracing and mouse modeling, we observed that platelet-derived growth factor receptor beta (PDGFRβ)+ pericytes give rise to developmental brown adipocytes but not to those in adult homeostasis. By contrast, T-box 18 (TBX18)+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRβ+ pericytes promotes brown adipogenesis by downregulating PDGFRβ. Furthermore, inhibition of Notch signaling in PDGFRβ+ pericytes mitigates high-fat, high-sucrose (HFHS)-induced glucose and metabolic impairment in mice during their development and juvenile phases. Collectively, these findings show that the Notch/PDGFRβ axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health.
Collapse
Affiliation(s)
- Zuoxiao Shi
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Shaolei Xiong
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ruoci Hu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zilai Wang
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jooman Park
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yanyu Qian
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jaden Wang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Pratibha Bhalla
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Nipun Velupally
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Minsun Stacey Jeon
- Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX 77030, USA
| | - Ke Kurt Zhang
- Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX 77030, USA
| | - Linlin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77845, USA
| | - Brian T Layden
- Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Jesse Brown Medical VA Medical Center, Chicago, IL 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA; Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
13
|
Zhao JY, Zhou LJ, Ma KL, Hao R, Li M. MHO or MUO? White adipose tissue remodeling. Obes Rev 2024; 25:e13691. [PMID: 38186200 DOI: 10.1111/obr.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 01/09/2024]
Abstract
In this review, we delve into the intricate relationship between white adipose tissue (WAT) remodeling and metabolic aspects in obesity, with a specific focus on individuals with metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO). WAT is a highly heterogeneous, plastic, and dynamically secreting endocrine and immune organ. WAT remodeling plays a crucial role in metabolic health, involving expansion mode, microenvironment, phenotype, and distribution. In individuals with MHO, WAT remodeling is beneficial, reducing ectopic fat deposition and insulin resistance (IR) through mechanisms like increased adipocyte hyperplasia, anti-inflammatory microenvironment, appropriate extracellular matrix (ECM) remodeling, appropriate vascularization, enhanced WAT browning, and subcutaneous adipose tissue (SWAT) deposition. Conversely, for those with MUO, WAT remodeling leads to ectopic fat deposition and IR, causing metabolic dysregulation. This process involves adipocyte hypertrophy, disrupted vascularization, heightened pro-inflammatory microenvironment, enhanced brown adipose tissue (BAT) whitening, and accumulation of visceral adipose tissue (VWAT) deposition. The review underscores the pivotal importance of intervening in WAT remodeling to hinder the transition from MHO to MUO. This insight is valuable for tailoring personalized and effective management strategies for patients with obesity in clinical practice.
Collapse
Affiliation(s)
- Jing Yi Zhao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Juan Zhou
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kai Le Ma
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Hao
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Li
- Research Laboratory of Molecular Biology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Li X, Sun D, Wang Z, Zhao Q, Liu Y, Hou Z. Transcriptional regulatory mechanism of NR2F2 and ZNF423 in avian preadipocyte differentiation. Gene 2024; 897:148106. [PMID: 38128789 DOI: 10.1016/j.gene.2023.148106] [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: 10/12/2023] [Revised: 12/09/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023]
Abstract
In the poultry industry, excessive abdominal fat deposition is not conducive to meat quality. Therefore, selection for optimal fat content levels in poultry has become a major breeding goal. We previously constructed NR2F2 overexpression (NR2F2OE) and knockout (NR2F2Δ/Δ/83-125aa) cell lines using Piggybac and CRISPR/Cas9 techniques, and confirmed that the transcription factor NR2F2 can significantly inhibit the differentiation of avian preadipocytes. In this study, we identified a downstream gene ZNF423 regulated by NR2F2, which is also involved in regulating avian fat deposition. First, we performed transcriptome analysis of the NR2F2-edited lines, which has been proven to be an inhibitor of avian fat deposition in our previous studies. Our findings revealed that NR2F2 affects a series of candidate regulators related to adipogenesis. Among these, we focused on ZNF423, which was significantly down-regulated in the NR2F2OE cell line and up-regulated in the NR2F2Δ/Δ/83-125aa cell line. Next, dual luciferase reporter assay results showed that the DNA-binding domain (DBDΔ72-143aa) of transcription factor NR2F2 may negatively affect the expression of downstream target gene ZNF423 by binding to its distal promoter region (-2356 to -2346). Moreover, we constructed a function analytical model and found that overexpression of ZNF423 significantly facilitated the differentiation of adipocytes in immortalized chicken preadipocytes (ICP1). Consistent with these findings, global transcriptome analysis of the ZNF423-overexpressed cell line (ZNF423OE) further demonstrated that the process of adipogenesis was significantly enriched. These results indicate that ZNF423 is a positive regulator of avian adipocyte differentiation. Overexpression of ZNF423 in the NR2F2OE cell line compensated for the inhibition of fat deposition phenotype, further suggesting that ZNF423 is a downstream target gene of NR2F2. These findings uncover a novel function of ZNF423 in avian adipocyte differentiation and analyzed the transcriptional regulation by its upstream transcription factor NR2F2. Additionally, we identified a list of functional candidate genes, providing important insights for further research on the mechanism of avian fat deposition.
Collapse
Affiliation(s)
- Xiaoqin Li
- National Engineering Laboratory for Animal Breeding and MARA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Dandan Sun
- National Engineering Laboratory for Animal Breeding and MARA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zheng Wang
- College of Life Sciences, Shanxi Agricultural University, Taiyuan 030801, China
| | - Qiangsen Zhao
- National Engineering Laboratory for Animal Breeding and MARA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yongtong Liu
- National Engineering Laboratory for Animal Breeding and MARA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhuocheng Hou
- National Engineering Laboratory for Animal Breeding and MARA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
15
|
Li B, Liu S, He Z, Luo E, Liu H. The role of zinc finger proteins in the fate determination of mesenchymal stem cells during osteogenic and adipogenic differentiation. Int J Biochem Cell Biol 2024; 167:106507. [PMID: 38142772 DOI: 10.1016/j.biocel.2023.106507] [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: 09/11/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
Zinc finger proteins (ZFPs) constitute a crucial group of transcription factors widely present in various organisms. They act as transcription factors, nucleases, and RNA-binding proteins, playing significant roles in cell differentiation, growth, and development. With extensive research on ZFPs, their roles in the determination of mesenchymal stem cells (MSCs) fate during osteogenic and adipogenic differentiation processes have become increasingly clear. ZFP521, for instance, is identified as an inhibitor of the Wnt signaling pathway and RUNX2's transcriptional activity, effectively suppressing osteogenic differentiation. Moreover, ZFP217 contributes to the inhibition of adipogenic differentiation by reducing the M6A level of the cell cycle regulator cyclin D1 (CCND1). In addition, other ZFPs can also influence the fate of mesenchymal stem cells (MSCs) during osteogenic and adipogenic differentiation through various signaling pathways, transcription factors, and epigenetic controls, participating in the subsequent differentiation and maturation of precursor cells. Given the prevalent occurrence of osteoporosis, obesity, and related metabolic disorders, a comprehensive understanding of the regulatory mechanisms balancing bone and fat metabolism is essential, with a particular focus on the fate determination of MSCs in osteogenic and adipogenic differentiation. In this review, we provide a detailed summary of how zinc finger proteins influence the osteogenic and adipogenic differentiation of MSCs through different signaling pathways, transcription factors, and epigenetic mechanisms. Additionally, we outline the regulatory mechanisms of ZFPs in controlling osteogenic and adipogenic differentiation based on various stages of MSC differentiation.
Collapse
Affiliation(s)
- Bolun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ze He
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
16
|
Benvie AM, Lee D, Jiang Y, Berry DC. Platelet-derived growth factor receptor beta is required for embryonic specification and confinement of the adult white adipose lineage. iScience 2024; 27:108682. [PMID: 38235323 PMCID: PMC10792241 DOI: 10.1016/j.isci.2023.108682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/31/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024] Open
Abstract
White adipose tissue (WAT) development and adult homeostasis rely on distinct adipocyte progenitor cells (APCs). While adult APCs are defined early during embryogenesis and generate adipocytes after WAT organogenesis, the mechanisms underlying adult adipose lineage determination and preservation remain undefined. Here, we uncover a critical role for platelet-derived growth factor receptor beta (Pdgfrβ) in identifying the adult APC lineage. Without Pdgfrβ, APCs lose their adipogenic competency to incite fibrotic tissue replacement and inflammation. Through lineage tracing analysis, we reveal that the adult APC lineage is lost and develops into macrophages when Pdgfrβ is deleted embryonically. Moreover, to maintain the APC lineage, Pdgfrβ activation stimulates p38/MAPK phosphorylation to promote APC proliferation and maintains the APC state by phosphorylating peroxisome proliferator activated receptor gamma (Pparγ) at serine 112. Together, our findings identify a role for Pdgfrβ acting as a rheostat for adult adipose lineage confinement to prevent unintended lineage switches.
Collapse
Affiliation(s)
- Abigail M. Benvie
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Derek Lee
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, University of Illinois College of Medicine at Chicago, Chicago, IL 60612, USA
| | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
17
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
18
|
Xining Z, Sai L. The Evolving Function of Vasculature and Pro-angiogenic Therapy in Fat Grafting. Cell Transplant 2024; 33:9636897241264976. [PMID: 39056562 PMCID: PMC11282510 DOI: 10.1177/09636897241264976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous fat grating is a widely-accepted method to correct soft tissue deficiency. Although fat transplantation shows excellent biocompatibility and simple applicability, the relatively low retention rate caused by fat necrosis is still a challenge. The vasculature is integral after fat grafting, serving multiple crucial functions. Rapid and effective angiogenesis within grafts is essential for supplying oxygen necessary for adipocytes' survival. It facilitates the influx of inflammatory cells to remove necrotic adipocytes and aids in the delivery of regenerative cells for adipose tissue regeneration in fat grafts. The vasculature also provides a niche for interaction between adipose progenitor cells and vascular progenitor cells, enhancing angiogenesis and adipogenesis in grafts. Various methods, such as enriching grafts with diverse pro-angiogenic cells or utilizing cell-free approaches, have been employed to enhance angiogenesis. Beige and dedifferentiated adipocytes in grafts could increase vessel density. This review aims to outline the function of vasculature in fat grafting and discuss different cell or cell-free approaches that can enhance angiogenesis following fat grafting.
Collapse
Affiliation(s)
- Zhang Xining
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Luo Sai
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
AlZaim I, de Rooij LPMH, Sheikh BN, Börgeson E, Kalucka J. The evolving functions of the vasculature in regulating adipose tissue biology in health and obesity. Nat Rev Endocrinol 2023; 19:691-707. [PMID: 37749386 DOI: 10.1038/s41574-023-00893-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 09/27/2023]
Abstract
Adipose tissue is an endocrine organ and a crucial regulator of energy storage and systemic metabolic homeostasis. Additionally, adipose tissue is a pivotal regulator of cardiovascular health and disease, mediated in part by the endocrine and paracrine secretion of several bioactive products, such as adipokines. Adipose vasculature has an instrumental role in the modulation of adipose tissue expansion, homeostasis and metabolism. The role of the adipose vasculature has been extensively explored in the context of obesity, which is recognized as a global health problem. Obesity-induced accumulation of fat, in combination with vascular rarefaction, promotes adipocyte dysfunction and induces oxidative stress, hypoxia and inflammation. It is now recognized that obesity-associated endothelial dysfunction often precedes the development of cardiovascular diseases. Investigations have revealed heterogeneity within the vascular niche and dynamic reciprocity between vascular and adipose cells, which can become dysregulated in obesity. Here we provide a comprehensive overview of the evolving functions of the vasculature in regulating adipose tissue biology in health and obesity.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Laura P M H de Rooij
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bilal N Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Emma Börgeson
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
20
|
Sabaratnam R, Hansen DR, Svenningsen P. White adipose tissue mitochondrial bioenergetics in metabolic diseases. Rev Endocr Metab Disord 2023; 24:1121-1133. [PMID: 37558853 DOI: 10.1007/s11154-023-09827-z] [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] [Accepted: 07/19/2023] [Indexed: 08/11/2023]
Abstract
White adipose tissue (WAT) is an important endocrine organ that regulates systemic energy metabolism. In metabolically unhealthy obesity, adipocytes become dysfunctional through hypertrophic mechanisms associated with a reduced endocrine function, reduced mitochondrial function, but increased inflammation, fibrosis, and extracellular remodelling. A pathologic WAT remodelling promotes systemic lipotoxicity characterized by fat accumulation in tissues such as muscle and liver, leading to systemic insulin resistance and type 2 diabetes. Several lines of evidence from human and animal studies suggest a link between unhealthy obesity and adipocyte mitochondrial dysfunction, and interventions that improve mitochondrial function may reduce the risk of obesity-associated diseases. This review discusses the importance of mitochondrial function and metabolism in human adipocyte biology and intercellular communication mechanisms within WAT. Moreover, a selected interventional approach for better adipocyte mitochondrial metabolism in humans is reviewed. A greater understanding of mitochondrial bioenergetics in WAT might provide novel therapeutic opportunities to prevent or restore dysfunctional adipose tissue in obesity-associated diseases.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Department of Clinical Research, University of Southern Denmark, Odense C, DK-5000, Denmark.
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark.
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| | - Didde Riisager Hansen
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| |
Collapse
|
21
|
Phan QM, Salz L, Kindl SS, Lopez JS, Thompson SM, Makkar J, Driskell IM, Driskell RR. Lineage commitment of dermal fibroblast progenitors is controlled by Kdm6b-mediated chromatin demethylation. EMBO J 2023; 42:e113880. [PMID: 37602956 PMCID: PMC10548174 DOI: 10.15252/embj.2023113880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
Dermal Fibroblast Progenitors (DFPs) differentiate into distinct fibroblast lineages during skin development. However, the epigenetic mechanisms that regulate DFP differentiation are not known. Our objective was to use multimodal single-cell approaches, epigenetic assays, and allografting techniques to define a DFP state and the mechanism that governs its differentiation potential. Our initial results indicated that the overall transcription profile of DFPs is repressed by H3K27me3 and has inaccessible chromatin at lineage-specific genes. Surprisingly, the repressive chromatin profile of DFPs renders them unable to reform the skin in allograft assays despite their multipotent potential. We hypothesized that chromatin derepression was modulated by the H3K27me3 demethylase, Kdm6b/Jmjd3. Dermal fibroblast-specific deletion of Kdm6b/Jmjd3 in mice resulted in adipocyte compartment ablation and inhibition of mature dermal papilla functions, confirmed by additional single-cell RNA-seq, ChIP-seq, and allografting assays. We conclude that DFPs are functionally derepressed during murine skin development by Kdm6b/Jmjd3. Our studies therefore reveal a multimodal understanding of how DFPs differentiate into distinct fibroblast lineages and provide a novel publicly available multiomics search tool.
Collapse
Affiliation(s)
- Quan M Phan
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Lucia Salz
- North Rhine‐Westphalia Technical University of AachenAachenGermany
| | - Sam S Kindl
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Jayden S Lopez
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Sean M Thompson
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Jasson Makkar
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Iwona M Driskell
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
| | - Ryan R Driskell
- School of Molecular BiosciencesWashington State UniversityPullmanWAUSA
- Center for Reproductive BiologyWashington State UniversityPullmanWAUSA
| |
Collapse
|
22
|
Santos MM, Costa TC, Silva W, Pistillo LZ, Junior DTV, Verardo LL, Paulino PVR, Sampaio CB, Gionbelli MP, Du M, Duarte MS. Nutrient supplementation of beef female calves at pre-weaning enhances the commitment of fibro-adipogenic progenitor cells to preadipocytes. Meat Sci 2023; 204:109286. [PMID: 37494740 DOI: 10.1016/j.meatsci.2023.109286] [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: 01/03/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/28/2023]
Abstract
We aimed to evaluate the impact of nutrient supplementation of beef female calves at pre-weaning on adipogenic determination. Thirty-four female calves were assigned to two experimental treatments: Control (CON, n = 17), where animals were supplemented only with mineral mixture; Supplemented (SUP, n = 17), where animals received energy-protein supplement containing minerals (5 g/kg of BW per day) of their body weight. Animals were supplemented from 100 to 250 days of age, and muscle samples were biopsied at the end of the supplementation period. Regarding the performance variables, there were no differences between treatments for initial body weight (P = 0.75). The final body weight (P = 0.07), average daily gain (P = 0.07), rib eye area (P = 0.03), and rib fat thickness (P = 0.08) were greater in SUP female calves compared with CON treatment. The number of fibro-adipogenic progenitor cells (P = 0.69) did not differ between treatments, while a greater number of intramuscular pre-adipocytes were observed in SUP than CON female calves (P = 0.01). The expression of miRNA-4429 (P = 0.20) did not differ between treatments, while the expression of miRNA-129-5p (P = 0.09) and miRNA-129-2-3p (P = 0.05) was greater in CON than SUP female calves. Our results suggest that nutrient supplementation at early postnatal stages of development enhances the commitment of fibro-adipogenic progenitor cells into the adipogenic lineages allowing to an increase in intramuscular fat deposition potential of the animals later in life.
Collapse
Affiliation(s)
- M M Santos
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, Viçosa, Brazil
| | - T C Costa
- Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Animal Science, Universidade Federal de Lavras, Lavras, MG, Brazil
| | - W Silva
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - L Z Pistillo
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
| | - D T Valente Junior
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - L L Verardo
- Department of Animal Science, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | | | - C B Sampaio
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
| | - M P Gionbelli
- Department of Animal Science, Universidade Federal de Lavras, Lavras, MG, Brazil
| | - M Du
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - M S Duarte
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
23
|
Ford H, Liu Q, Fu X, Strieder-Barboza C. White Adipose Tissue Heterogeneity in the Single-Cell Era: From Mice and Humans to Cattle. BIOLOGY 2023; 12:1289. [PMID: 37886999 PMCID: PMC10604679 DOI: 10.3390/biology12101289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023]
Abstract
Adipose tissue is a major modulator of metabolic function by regulating energy storage and by acting as an endocrine organ through the secretion of adipokines. With the advantage of next-generation sequencing-based single-cell technologies, adipose tissue has been studied at single-cell resolution, thus providing unbiased insight into its molecular composition. Recent single-cell RNA sequencing studies in human and mouse models have dissected the transcriptional cellular heterogeneity of subcutaneous (SAT), visceral (VAT), and intramuscular (IMAT) white adipose tissue depots and revealed unique populations of adipose tissue progenitor cells, mature adipocytes, immune cell, vascular cells, and mesothelial cells that play direct roles on adipose tissue function and the development of metabolic disorders. In livestock species, especially in bovine, significant gaps of knowledge remain in elucidating the roles of adipose tissue cell types and depots on driving the pathogenesis of metabolic disorders and the distinct fat deposition in VAT, SAT, and IMAT in meat animals. This review summarizes the current knowledge on the transcriptional and functional cellular diversity of white adipose tissue revealed by single-cell approaches and highlights the depot-specific function of adipose tissue in different mammalian species, with a particular focus on recent findings and future implications in cattle.
Collapse
Affiliation(s)
- Hunter Ford
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409, USA;
| | - Qianglin Liu
- School of Animal Sciences, Agricultural Center, Louisiana State University, Baton Rouge, LA 70803, USA; (Q.L.); (X.F.)
| | - Xing Fu
- School of Animal Sciences, Agricultural Center, Louisiana State University, Baton Rouge, LA 70803, USA; (Q.L.); (X.F.)
| | - Clarissa Strieder-Barboza
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409, USA;
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX 79106, USA
| |
Collapse
|
24
|
Lee DK, Kim M, Jeong J, Lee YS, Yoon JW, An MJ, Jung HY, Kim CH, Ahn Y, Choi KH, Jo C, Lee CK. Unlocking the potential of stem cells: Their crucial role in the production of cultivated meat. Curr Res Food Sci 2023; 7:100551. [PMID: 37575132 PMCID: PMC10412782 DOI: 10.1016/j.crfs.2023.100551] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Cellular agriculture is an emerging research field of agribiotechnology that aims to produce agricultural products using stem cells, without sacrificing animals or cultivating crops. Cultivated meat, as a representative cellular product of cellular agriculture, is being actively researched due to global food insecurity, environmental, and ethical concerns. This review focuses on the application of stem cells, which are the seeds of cellular agriculture, for the production of cultivated meat, with emphasis on deriving and culturing muscle and adipose stem cells for imitating fresh meat. Establishing standards and safety regulations for culturing stem cells is crucial for the market entry of cultured muscle tissue-based biomaterials. Understanding stem cells is a prerequisite for creating reliable cultivated meat and other cellular agricultural biomaterials. The techniques and regulations from the cultivated meat industry could pave the way for new cellular agriculture industries in the future.
Collapse
Affiliation(s)
- Dong-Kyung Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Minsu Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young-Seok Lee
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Ji Won Yoon
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Min-Jeong An
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Hyun Young Jung
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cho Hyun Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yelim Ahn
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwang-Hwan Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Research and Development Center, Space F Corporation, Hwasung, 18471, Gyeonggi-do, Republic of Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Gangwon-do, Republic of Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Gangwon-do, Republic of Korea
| |
Collapse
|
25
|
Wang B, Du M. Increasing adipocyte number and reducing adipocyte size: the role of retinoids in adipose tissue development and metabolism. Crit Rev Food Sci Nutr 2023; 64:10608-10625. [PMID: 37427553 PMCID: PMC10776826 DOI: 10.1080/10408398.2023.2227258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The rising prevalence of obesity is a grave public health threat. In response to excessive energy intake, adipocyte hypertrophy impairs cellular function and leads to metabolic dysfunctions while de novo adipogenesis leads to healthy adipose tissue expansion. Through burning fatty acids and glucose, the thermogenic activity of brown/beige adipocytes can effectively reduce the size of adipocytes. Recent studies show that retinoids, especially retinoic acid (RA), promote adipose vascular development which in turn increases the number of adipose progenitors surrounding the vascular vessels. RA also promotes preadipocyte commitment. In addition, RA promotes white adipocyte browning and stimulates the thermogenic activity of brown/beige adipocytes. Thus, vitamin A is a promising anti-obesity micronutrient.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Min Du
- Laboratory of Nutrigenomics and Growth Biology, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
26
|
Rivera-Gonzalez GC, Butka EG, Gonzalez CE, Kong W, Jindal K, Morris SA. Single-cell lineage tracing reveals hierarchy and mechanism of adipocyte precursor maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543318. [PMID: 37398135 PMCID: PMC10312565 DOI: 10.1101/2023.06.01.543318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
White adipose tissue is crucial in various physiological processes. In response to high caloric intake, adipose tissue may expand by generating new adipocytes. Adipocyte precursor cells (progenitors and preadipocytes) are essential for generating mature adipocytes, and single-cell RNA sequencing provides new means to identify these populations. Here, we characterized adipocyte precursor populations in the skin, an adipose depot with rapid and robust generation of mature adipocytes. We identified a new population of immature preadipocytes, revealed a biased differentiation potential of progenitor cells, and identified Sox9 as a critical factor in driving progenitors toward adipose commitment, the first known mechanism of progenitor differentiation. These findings shed light on the specific dynamics and molecular mechanisms underlying rapid adipogenesis in the skin.
Collapse
Affiliation(s)
- Guillermo C. Rivera-Gonzalez
- Department of Developmental Biology, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Emily G. Butka
- Department of Developmental Biology, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Carolynn E. Gonzalez
- Department of Developmental Biology, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Wenjun Kong
- Department of Developmental Biology, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Kunal Jindal
- Department of Developmental Biology, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Samantha A. Morris
- Department of Developmental Biology, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine; 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
27
|
Abstract
The circadian clock plays an essential role in coordinating feeding and metabolic rhythms with the light/dark cycle. Disruption of clocks is associated with increased adiposity and metabolic disorders, whereas aligning feeding time with cell-autonomous rhythms in metabolism improves health. Here, we provide a comprehensive overview of recent literature in adipose tissue biology as well as our understanding of molecular mechanisms underlying the circadian regulation of transcription, metabolism, and inflammation in adipose tissue. We highlight recent efforts to uncover the mechanistic links between clocks and adipocyte metabolism, as well as its application to dietary and behavioral interventions to improve health and mitigate obesity.
Collapse
Affiliation(s)
- Chelsea Hepler
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
28
|
Shi Z, Xiong S, Hu R, Wang Z, Park J, Qian Y, Wang J, Bhalla P, Velupally N, Song Q, Song Z, Layden BT, Jiang Y. The Notch-Pdgfrβ axis suppresses brown adipocyte progenitor differentiation in early postnatal mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.541839. [PMID: 37293108 PMCID: PMC10245810 DOI: 10.1101/2023.05.24.541839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively studied. Here through in vivo lineage tracing, we observed that PDGFRβ+ pericytes give rise to developmental brown adipocytes, but not to those in adult homeostasis. In contrast, TBX18+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRβ+ pericytes promotes brown adipogenesis through the downregulation of PDGFRβ. Furthermore, inhibition of Notch signaling in PDGFRβ+ pericytes mitigates HFHS (high-fat, high-sucrose) induced glucose and metabolic impairment in both developmental and adult stages. Collectively, these findings show that the Notch/PDGFRβ axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health. Highlights PDGFRβ+ pericytes act as an essential developmental brown APC.TBX18+ pericytes contribute to brown adipogenesis in a depot-specific manner.Inhibiting Notch-Pdgfrβ axis promotes brown APC adipogenesis.Enhanced postnatal brown adipogenesis improves metabolic health in adult stage.
Collapse
|
29
|
Jääskeläinen I, Petäistö T, Mirzarazi Dahagi E, Mahmoodi M, Pihlajaniemi T, Kaartinen MT, Heljasvaara R. Collagens Regulating Adipose Tissue Formation and Functions. Biomedicines 2023; 11:biomedicines11051412. [PMID: 37239083 DOI: 10.3390/biomedicines11051412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
The globally increasing prevalence of obesity is associated with the development of metabolic diseases such as type 2 diabetes, dyslipidemia, and fatty liver. Excess adipose tissue (AT) often leads to its malfunction and to a systemic metabolic dysfunction because, in addition to storing lipids, AT is an active endocrine system. Adipocytes are embedded in a unique extracellular matrix (ECM), which provides structural support to the cells as well as participating in the regulation of their functions, such as proliferation and differentiation. Adipocytes have a thin pericellular layer of a specialized ECM, referred to as the basement membrane (BM), which is an important functional unit that lies between cells and tissue stroma. Collagens form a major group of proteins in the ECM, and some of them, especially the BM-associated collagens, support AT functions and participate in the regulation of adipocyte differentiation. In pathological conditions such as obesity, AT often proceeds to fibrosis, characterized by the accumulation of large collagen bundles, which disturbs the natural functions of the AT. In this review, we summarize the current knowledge on the vertebrate collagens that are important for AT development and function and include basic information on some other important ECM components, principally fibronectin, of the AT. We also briefly discuss the function of AT collagens in certain metabolic diseases in which they have been shown to play central roles.
Collapse
Affiliation(s)
- Iida Jääskeläinen
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Tiina Petäistö
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Elahe Mirzarazi Dahagi
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Mahdokht Mahmoodi
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Taina Pihlajaniemi
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| | - Mari T Kaartinen
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| | - Ritva Heljasvaara
- ECM-Hypoxia Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90014 Oulu, Finland
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Phan QM, Salz L, Kindl SS, Lopez JS, Thompson SM, Makkar J, Driskell IM, Driskell RR. Lineage Commitment of Dermal Fibroblast Progenitors is Mediated by Chromatin De-repression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531478. [PMID: 36945417 PMCID: PMC10028926 DOI: 10.1101/2023.03.07.531478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Dermal Fibroblast Progenitors (DFPs) differentiate into distinct fibroblast lineages during skin development. However, the mechanisms that regulate lineage commitment of naive dermal progenitors to form niches around the hair follicle, dermis, and hypodermis, are unknown. In our study, we used multimodal single-cell approaches, epigenetic assays, and allografting techniques to define a DFP state and the mechanisms that govern its differentiation potential. Our results indicate that the overall chromatin profile of DFPs is repressed by H3K27me3 and has inaccessible chromatin at lineage specific genes. Surprisingly, the repressed chromatin profile of DFPs renders them unable to reform skin in allograft assays despite their multipotent potential. Distinct fibroblast lineages, such as the dermal papilla and adipocytes contained specific chromatin profiles that were de-repressed during late embryogenesis by the H3K27-me3 demethylase, Kdm6b/Jmjd3. Tissue-specific deletion of Kdm6b/Jmjd3 resulted in ablating the adipocyte compartment and inhibiting mature dermal papilla functions in single-cell-RNA-seq, ChIPseq, and allografting assays. Altogether our studies reveal a mechanistic multimodal understanding of how DFPs differentiate into distinct fibroblast lineages, and we provide a novel multiomic search-tool within skinregeneration.org.
Collapse
Affiliation(s)
- Quan M. Phan
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Lucia Salz
- North Rhine-Westphalia Technical University of Aachen, Aachen, Germany
| | - Sam S. Kindl
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Jayden S. Lopez
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Sean M. Thompson
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Jasson Makkar
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Iwona M. Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA
| | - Ryan R. Driskell
- School of Molecular Biosciences, Washington State University, Pullman, WA
- Center for Reproductive Biology, Washington State University, Pullman, WA
| |
Collapse
|
33
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
34
|
Roth L, Johann K, Hönes GS, Oelkrug R, Wagner L, Hoffmann A, Krohn K, Moeller LC, Weiner J, Heiker JT, Klöting N, Tönjes A, Stumvoll M, Blüher M, Mittag J, Krause K. Thyroid hormones regulate Zfp423 expression in regionally distinct adipose depots through direct and cell-autonomous action. Cell Rep 2023; 42:112088. [PMID: 36753417 DOI: 10.1016/j.celrep.2023.112088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 12/05/2022] [Accepted: 01/24/2023] [Indexed: 02/09/2023] Open
Abstract
The hypothalamic pituitary thyroid axis is a major regulator of many differentiation processes, including adipose tissue. However, it remains unclear whether and how thyroid hormone (TH) signaling contributes to preadipocyte commitment and differentiation into mature adipocytes. Here, we show a cell-autonomous effect of TH on the transcriptional regulation of zinc finger protein 423 (Zfp423), an early adipogenic determination factor, in murine adipose depots. Mechanistically, binding of the unliganded TH receptor to a negative TH responsive element within the Zfp423 promoter activates transcriptional activity that is reversed upon TH binding. Zfp423 upregulation is associated with increased GFP+ preadipocyte recruitment in stromal vascular fraction isolated from white fat of hypothyroid Zfp423GFP reporter mice. RNA sequencing identified Zfp423-driven gene programs that are modulated in response to TH during adipogenic differentiation. Collectively, we identified Zfp423 as a key molecule that integrates TH signaling into the regulation of adipose tissue plasticity.
Collapse
Affiliation(s)
- Lisa Roth
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Kornelia Johann
- Institute for Endocrinology and Diabetes/Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Georg Sebastian Hönes
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Rebecca Oelkrug
- Institute for Endocrinology and Diabetes/Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Leonie Wagner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Knut Krohn
- DNA Core Unit Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Lars C Moeller
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Juliane Weiner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - John T Heiker
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Anke Tönjes
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Michael Stumvoll
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Matthias Blüher
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Jens Mittag
- Institute for Endocrinology and Diabetes/Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Deutsches Zentrum für Diabetesforschung e.V., 85764 Neuherberg, Germany.
| |
Collapse
|
35
|
Abstract
Rather than serving as a mere onlooker, adipose tissue is a complex endocrine organ and active participant in disease initiation and progression. Disruptions of biological processes operating within adipose can disturb healthy systemic physiology, the sequelae of which include metabolic disorders such as obesity and type 2 diabetes. A burgeoning interest in the field of adipose research has allowed for the elucidation of regulatory networks underlying both adipose tissue function and dysfunction. Despite this progress, few diseases are treated by targeting maladaptation in the adipose, an oft-overlooked organ. In this review, we elaborate on the distinct subtypes of adipocytes, their developmental origins and secretory roles, and the dynamic interplay at work within the tissue itself. Central to this discussion is the relationship between adipose and disease states, including obesity, cachexia, and infectious diseases, as we aim to leverage our wealth of knowledge for the development of novel and targeted therapeutics.
Collapse
Affiliation(s)
- Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; .,Howard Hughes Medical Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
36
|
Burkhardt LM, Bucher CH, Löffler J, Rinne C, Duda GN, Geissler S, Schulz TJ, Schmidt-Bleek K. The benefits of adipocyte metabolism in bone health and regeneration. Front Cell Dev Biol 2023; 11:1104709. [PMID: 36895792 PMCID: PMC9988968 DOI: 10.3389/fcell.2023.1104709] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Patients suffering from musculoskeletal diseases must cope with a diminished quality of life and an increased burden on medical expenses. The interaction of immune cells and mesenchymal stromal cells during bone regeneration is one of the key requirements for the restoration of skeletal integrity. While stromal cells of the osteo-chondral lineage support bone regeneration, an excessive accumulation of cells of the adipogenic lineage is thought to promote low-grade inflammation and impair bone regeneration. Increasing evidence indicates that pro-inflammatory signaling from adipocytes is responsible for various chronic musculoskeletal diseases. This review aims to summarize the features of bone marrow adipocytes by phenotype, function, secretory features, metabolic properties and their impact on bone formation. In detail, the master regulator of adipogenesis and prominent diabetes drug target, peroxisome proliferator-activated receptor γ (PPARG), will be debated as a potential therapeutic approach to enhance bone regeneration. We will explore the possibilities of using clinically established PPARG agonists, the thiazolidinediones (TZDs), as a treatment strategy to guide the induction of a pro-regenerative, metabolically active bone marrow adipose tissue. The impact of this PPARG induced bone marrow adipose tissue type on providing the necessary metabolites to sustain osteogenic-as well as beneficial immune cells during bone fracture healing will be highlighted.
Collapse
Affiliation(s)
- Lisa-Marie Burkhardt
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Julia Löffler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Charlotte Rinne
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
37
|
Scamfer SR, Lee MD, Hilgendorf KI. Ciliary control of adipocyte progenitor cell fate regulates energy storage. Front Cell Dev Biol 2022; 10:1083372. [PMID: 36561368 PMCID: PMC9763467 DOI: 10.3389/fcell.2022.1083372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The primary cilium is a cellular sensory organelle found in most cells in our body. This includes adipocyte progenitor cells in our adipose tissue, a complex organ involved in energy storage, endocrine signaling, and thermogenesis. Numerous studies have shown that the primary cilium plays a critical role in directing the cell fate of adipocyte progenitor cells in multiple adipose tissue types. Accordingly, diseases with dysfunctional cilia called ciliopathies have a broad range of clinical manifestations, including obesity and diabetes. This review summarizes our current understanding of how the primary cilium regulates adipocyte progenitor cell fate in multiple contexts and illustrates the importance of the primary cilium in regulating energy storage and adipose tissue function.
Collapse
Affiliation(s)
| | | | - Keren I. Hilgendorf
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
38
|
Fischer AW, Jaeckstein MY, Heeren J. Lysosomal acid lipase promotes endothelial proliferation in cold-activated adipose tissue. Adipocyte 2022; 11:28-33. [PMID: 34957913 PMCID: PMC8726628 DOI: 10.1080/21623945.2021.2013416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Oxidative tissues such as brown adipose tissue and muscle internalize large amounts of circulating lipids and glucose as energy source. Endothelial cells (ECs) provide a platform for regulated transport and processing of blood-borne nutrients. Next to this role, it has become recognized that intercellular crosstalk between ECs and underlying parenchymal cells is indispensable for maintenance of tissue homoeostasis. Here, we comment on our recent observation that capillary ECs in thermogenic adipose tissues take up and metabolize entire triglyceride-rich lipoprotein (TRL) particles in response to cold exposure. This process is dependent on CD36, lipoprotein lipase (LPL) and lysosomal acid lipase (LAL). Remarkably, loss of LAL specifically in endothelial cells results in impaired endothelial proliferation and diminished thermogenic adaptation. Mechanistically, cell culture experiments indicate that LAL-mediated TRL processing leads to the generation of reactive oxygen species, which in turn activate hypoxia-induced factor (HIF)-mediated proliferative responses. In the current manuscript, we provide in vivo evidence that LAL-deficiency impairs proliferation of endothelial cells in thermogenic adipose tissue. In addition, we show uptake of nanoparticle-labelled TRL and LAL expression in cardiac endothelial cells, suggesting a physiological function of endothelial lipoprotein processing not only in thermogenic adipose tissue but also in cardiac muscle.
Collapse
Affiliation(s)
- Alexander W. Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michelle Y. Jaeckstein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
39
|
Li Q, Spalding KL. The regulation of adipocyte growth in white adipose tissue. Front Cell Dev Biol 2022; 10:1003219. [PMID: 36483678 PMCID: PMC9723158 DOI: 10.3389/fcell.2022.1003219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/03/2022] [Indexed: 10/25/2023] Open
Abstract
Adipocytes can increase in volume up to a thousand-fold, storing excess calories as triacylglycerol in large lipid droplets. The dramatic morphological changes required of adipocytes demands extensive cytoskeletal remodeling, including lipid droplet and plasma membrane expansion. Cell growth-related signalling pathways are activated, stimulating the production of sufficient amino acids, functional lipids and nucleotides to meet the increasing cellular needs of lipid storage, metabolic activity and adipokine secretion. Continued expansion gives rise to enlarged (hypertrophic) adipocytes. This can result in a failure to maintain growth-related homeostasis and an inability to cope with excess nutrition or respond to stimuli efficiently, ultimately leading to metabolic dysfunction. We summarize recent studies which investigate the functional and cellular structure remodeling of hypertrophic adipocytes. How adipocytes adapt to an enlarged cell size and how this relates to cellular dysfunction are discussed. Understanding the healthy and pathological processes involved in adipocyte hypertrophy may shed light on new strategies for promoting healthy adipose tissue expansion.
Collapse
Affiliation(s)
- Qian Li
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Kirsty L. Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Nurlaila I, Roh K, Yeom CH, Kang H, Lee S. Acquired lymphedema: Molecular contributors and future directions for developing intervention strategies. Front Pharmacol 2022; 13:873650. [PMID: 36386144 PMCID: PMC9640931 DOI: 10.3389/fphar.2022.873650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/13/2022] [Indexed: 08/05/2023] Open
Abstract
Lymphedema is a debilitating chronic disease that mostly develops as an adverse reaction to cancer treatment modalities such as chemotherapy, surgery, and radiotherapy. Lymphedema also appears to be a deteriorating consequence of roundworm infections, as best represented by filariasis. According to its origin, lymphedema is classified as primary lymphedema and acquired lymphedema. The latter is an acquired condition that, hitherto, received a considerably low attention owing to the less number of fatal cases been reported. Notably, despite the low mortality rate in lymphedema, it has been widely reported to reduce the disease-free survival and thus the quality of life of affected patients. Hence, in this review, we focused on acquired lymphedema and orchestration of molecular interplays associated with either stimulation or inhibition of lymphedema development that were, in vast majority, clearly depicted in animal models with their specific and distinct technical approaches. We also discussed some recent progress made in phytochemical-based anti-lymphedema intervention strategies and the specific mechanisms underlying their anti-lymphedema properties. This review is crucial to understand not only the comprehensive aspects of the disease but also the future directions of the intervention strategies that can address the quality of life of affected patients rather than alleviating apparent symptoms only.
Collapse
Affiliation(s)
- Ika Nurlaila
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
- Department of Vaccine and Drugs, The National Research and Innovation Agency, Jakarta, Indonesia
| | - Kangsan Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Division of Cardiology and Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Hee Kang
- Humanitas College, Kyung Hee University, Yongin, South Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
41
|
Costa T, Lourenço P, Souza R, Lopes M, Araújo R, Santos M, Luciano L, Massensini J, Chalfun L, Rennó L, Sampaio C, Veroneze R, Paulino P, Gionbelli M, Duarte M. Ruminal undegradable protein enriched diet during late gestation of beef cows affects maternal metabolism and offspring’s skeletal muscle development. Anim Feed Sci Technol 2022. [DOI: 10.1016/j.anifeedsci.2022.115400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Zhang Q, Shan B, Guo L, Shao M, Vishvanath L, Elmquist G, Xu L, Gupta RK. Distinct functional properties of murine perinatal and adult adipose progenitor subpopulations. Nat Metab 2022; 4:1055-1070. [PMID: 35982290 PMCID: PMC9940036 DOI: 10.1038/s42255-022-00613-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 07/06/2022] [Indexed: 01/25/2023]
Abstract
Adult white adipose tissue (WAT) harbors distinct mesenchymal stromal cell subpopulations that differentially affect WAT function and plasticity. Here we unveil the cellular landscape of the perinatal epididymal WAT primordium using single-cell transcriptomics in male mice. We reveal that adipocyte precursor cells and fibro-inflammatory progenitors (FIPs) emerge as functionally distinct PDGFRβ+ subpopulations within the epididymal WAT anlagen prior to adipocyte accrual. We further identify important molecular and functional differences between perinatal and adult FIPs, including differences in their pro-inflammatory response, adipogenic capacity and anti-adipogenic behavior. Notably, we find that transient overexpression of Pparg in PDGFRβ+ cells only during postnatal days 0.5 to 7.5 in male mice leads to hyperplastic WAT development, durable progenitor cell reprogramming, and protection against pathologic WAT remodeling and glucose intolerance in adult-onset obesity. Thus, factors that alter the adipogenic capacity of perinatal adipose progenitors can have long-lasting effects on progenitor plasticity, tissue expandability and metabolic health into adulthood.
Collapse
Affiliation(s)
- Qianbin Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bo Shan
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lavanya Vishvanath
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Elmquist
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, 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
|
43
|
Lendahl U, Muhl L, Betsholtz C. Identification, discrimination and heterogeneity of fibroblasts. Nat Commun 2022; 13:3409. [PMID: 35701396 PMCID: PMC9192344 DOI: 10.1038/s41467-022-30633-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 05/04/2022] [Indexed: 12/14/2022] Open
Abstract
Fibroblasts, the principal cell type of connective tissue, secrete extracellular matrix components during tissue development, homeostasis, repair and disease. Despite this crucial role, the identification and distinction of fibroblasts from other cell types are challenging and laden with caveats. Rapid progress in single-cell transcriptomics now yields detailed molecular portraits of fibroblasts and other cell types in our bodies, which complement and enrich classical histological and immunological descriptions, improve cell class definitions and guide further studies on the functional heterogeneity of cell subtypes and states, origins and fates in physiological and pathological processes. In this review, we summarize and discuss recent advances in the understanding of fibroblast identification and heterogeneity and how they discriminate from other cell types. In this review, the authors look at how recent progress in single-cell transcriptomics complement and enrich the classical, largely morphological, portraits of fibroblasts. The detailed molecular information now available provides new insights into fibroblast identity, heterogeneity and function.
Collapse
Affiliation(s)
- Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77, Stockholm, Sweden.,Department of Neurobiology, Care sciences and Society, Karolinska Institutet, SE-14183, Huddinge, Sweden
| | - Lars Muhl
- Department of Medicine, Huddinge, Karolinska Institutet, Blickagången 16, SE-141 57, Huddinge, Sweden
| | - Christer Betsholtz
- Department of Medicine, Huddinge, Karolinska Institutet, Blickagången 16, SE-141 57, Huddinge, Sweden. .,Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
44
|
Steiner BM, Berry DC. The Regulation of Adipose Tissue Health by Estrogens. Front Endocrinol (Lausanne) 2022; 13:889923. [PMID: 35721736 PMCID: PMC9204494 DOI: 10.3389/fendo.2022.889923] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 12/14/2022] Open
Abstract
Obesity and its' associated metabolic diseases such as type 2 diabetes and cardiometabolic disorders are significant health problems confronting many countries. A major driver for developing obesity and metabolic dysfunction is the uncontrolled expansion of white adipose tissue (WAT). Specifically, the pathophysiological expansion of visceral WAT is often associated with metabolic dysfunction due to changes in adipokine secretion profiles, reduced vascularization, increased fibrosis, and enrichment of pro-inflammatory immune cells. A critical determinate of body fat distribution and WAT health is the sex steroid estrogen. The bioavailability of estrogen appears to favor metabolically healthy subcutaneous fat over visceral fat growth while protecting against changes in metabolic dysfunction. Our review will focus on the role of estrogen on body fat partitioning, WAT homeostasis, adipogenesis, adipocyte progenitor cell (APC) function, and thermogenesis to control WAT health and systemic metabolism.
Collapse
Affiliation(s)
| | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
45
|
Strohmeier K, Hofmann M, Jacak J, Narzt MS, Wahlmueller M, Mairhofer M, Schaedl B, Holnthoner W, Barsch M, Sandhofer M, Wolbank S, Priglinger E. Multi-Level Analysis of Adipose Tissue Reveals the Relevance of Perivascular Subpopulations and an Increased Endothelial Permeability in Early-Stage Lipedema. Biomedicines 2022; 10:biomedicines10051163. [PMID: 35625899 PMCID: PMC9138324 DOI: 10.3390/biomedicines10051163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Lipedema is a chronic, progressive disease of adipose tissue with unknown etiology. Based on the relevance of the stromal vascular fraction (SVF) cell population in lipedema, we performed a thorough characterization of subcutaneous adipose tissue, SVF isolated thereof and the sorted populations of endothelial cells (EC), pericytes and cultured adipose-derived stromal/stem cells (ASC) of early-stage lipedema patients. We employed histological and gene expression analysis and investigated the endothelial barrier by immunofluorescence and analysis of endothelial permeability in vitro. Although there were no significant differences in histological stainings, we found altered gene expression of factors relevant for local estrogen metabolism (aromatase), preadipocyte commitment (ZNF423) and immune cell infiltration (CD11c) in lipedema on the tissue level, as well as in distinct cellular subpopulations. Machine learning analysis of immunofluorescence images of CD31 and ZO-1 revealed a morphological difference in the cellular junctions of EC cultures derived from healthy and lipedema individuals. Furthermore, the secretome of lipedema-derived SVF cells was sufficient to significantly increase leakiness of healthy human primary EC, which was also reflected by decreased mRNA expression of VE-cadherin. Here, we showed for the first time that the secretome of SVF cells creates an environment that triggers endothelial barrier dysfunction in early-stage lipedema. Moreover, since alterations in gene expression were detected on the cellular and/or tissue level, the choice of sample material is of high importance in elucidating this complex disease.
Collapse
Affiliation(s)
- Karin Strohmeier
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria; (K.S.); (M.-S.N.); (M.W.); (B.S.); (W.H.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
| | - Martina Hofmann
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
- School of Medical Engineering and Applied Social Science, University of Applied Sciences Upper Austria, 4020 Linz, Austria
| | - Jaroslaw Jacak
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
- School of Medical Engineering and Applied Social Science, University of Applied Sciences Upper Austria, 4020 Linz, Austria
| | - Marie-Sophie Narzt
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria; (K.S.); (M.-S.N.); (M.W.); (B.S.); (W.H.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
| | - Marlene Wahlmueller
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria; (K.S.); (M.-S.N.); (M.W.); (B.S.); (W.H.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
| | - Mario Mairhofer
- Department of Hematology and Internal Oncology, Johannes Kepler University, 4020 Linz, Austria;
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria; (K.S.); (M.-S.N.); (M.W.); (B.S.); (W.H.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
- University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfgang Holnthoner
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria; (K.S.); (M.-S.N.); (M.W.); (B.S.); (W.H.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
| | - Martin Barsch
- Austrian Center for Lipedema, Skin Aesthetic Vein Laser Practice, 4020 Linz, Austria; (M.B.); (M.S.)
| | - Matthias Sandhofer
- Austrian Center for Lipedema, Skin Aesthetic Vein Laser Practice, 4020 Linz, Austria; (M.B.); (M.S.)
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria; (K.S.); (M.-S.N.); (M.W.); (B.S.); (W.H.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
| | - Eleni Priglinger
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria; (K.S.); (M.-S.N.); (M.W.); (B.S.); (W.H.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; (M.H.); (J.J.)
- Correspondence:
| |
Collapse
|
46
|
Xue C, Li X, Ba L, Shen Y, Sun Z, Gu J, Yang Y, Han Q, Zhao RC. Irisin mediates beiging of adipose-derived mesenchymal stem cells through binding to TRPC3. BMC Biol 2022; 20:95. [PMID: 35501783 PMCID: PMC9063202 DOI: 10.1186/s12915-022-01287-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Beiging of white fat plays an important role in energy metabolism. Beige adipocytes contribute to the regulation of body weight and body temperature through expenditure of chemical energy to produce heat, and they have therefore recently attracted considerable attention as potential targets for therapeutic approaches in metabolic disorders, including obesity. All adipocytes, including beige adipocytes, differentiate from mesenchymal stem cells (MSCs), which may provide an important path for clinical intervention; however, the mechanism of beiging of human adipose cell-derived MSCs is not fully understood. Here, we provide insights on the role of IRISIN, which is known to be secreted by skeletal muscle and promote beiging of white fat. RESULTS We established an IRISIN-induced mesenchymal stem cell beiging model and found that IRISIN protein interacts with the MSC membrane protein TRPC3. This interaction results in calcium influx and consequential activation of Erk and Akt signaling pathways, which causes phosphorylation of PPARγ. The phosphorylated PPARγ enters the nucleus and binds the UCP1 promoter region. Furthermore, the role of TRPC3 in the beiging of MSCs was largely abolished in Trpc3-/- mice. We additionally demonstrate that the calcium concentration in the brain of mice increases upon IRISIN stimulation, followed by an increase in the content of excitatory amino acids and norepinephrine, while Trpc3-/- mice exhibit the reverse effect. CONCLUSIONS We found that TRPC3 is a key factor in irisin-induced beiging of MSCs, which may provide a new target pathway in addressing metabolic disorders. Our results additionally suggest that the interaction of irisin with TRPC3 may affect multiple tissues, including the brain.
Collapse
Affiliation(s)
- Chunling Xue
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China.
| | - Xuechun Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China
| | - Li Ba
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China
| | - Yamei Shen
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China
| | - Zhao Sun
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Junjie Gu
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Ying Yang
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, No. 1 Shuaifuyuan Hutong, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory (No.BZO381), Beijing, China.
| |
Collapse
|
47
|
Neonatal vitamin A administration increases intramuscular fat by promoting angiogenesis and preadipocyte formation. Meat Sci 2022; 191:108847. [DOI: 10.1016/j.meatsci.2022.108847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022]
|
48
|
Dang TN, Tiongco RP, Brown LM, Taylor JL, Lyons JM, Lau FH, Floyd ZE. Expression of the preadipocyte marker ZFP423 is dysregulated between well-differentiated and dedifferentiated liposarcoma. BMC Cancer 2022; 22:300. [PMID: 35313831 PMCID: PMC8939188 DOI: 10.1186/s12885-022-09379-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/04/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Well-differentiated and dedifferentiated liposarcomas are rare soft tissue tumors originating in adipose tissue that share genetic abnormalities but have significantly different metastatic potential. Dedifferentiated liposarcoma (DDLPS) is highly aggressive and has an overall 5-year survival rate of 30% as compared to 90% for well-differentiated liposarcoma (WDLPS). This discrepancy may be connected to their potential to form adipocytes, where WDLPS is adipogenic but DDLPS is adipogenic-impaired. Normal adipogenesis requires Zinc Finger Protein 423 (ZFP423), a transcriptional coregulator of Perixosome Proliferator Activated Receptor gamma (PPARG2) mRNA expression that defines committed preadipocytes. Expression of ZFP423 in preadipocytes is promoted by Seven-In-Absentia Homolog 2 (SIAH2)-mediated degradation of Zinc Finger Protein 521 (ZFP521). This study investigated the potential role of ZFP423, SIAH2 and ZFP521 in the adipogenic potential of WDLPS and DDLPS. METHODS Human WDLPS and DDLPS fresh and paraffin-embedded tissues were used to assess the gene and protein expression of proadipogenic regulators. In parallel, normal adipose tissue stromal cells along with WDLPS and DDLPS cell lines were cultured, genetically modified, and induced to undergo adipogenesis in vitro. RESULTS Impaired adipogenic potential in DDLPS was associated with reduced ZFP423 protein levels in parallel with reduced PPARG2 expression, potentially involving regulation of ZFP521. SIAH2 protein levels did not define a clear distinction related to adipogenesis in these liposarcomas. However, in primary tumor specimens, SIAH2 mRNA was consistently upregulated in DDLPS compared to WDLPS when assayed by fluorescence in situ hybridization or real-time PCR. CONCLUSIONS These data provide novel insights into ZFP423 expression in adipogenic regulation between WDLPS and DDLPS adipocytic tumor development. The data also introduces SIAH2 mRNA levels as a possible molecular marker to distinguish between WDLPS and DDLPS.
Collapse
Affiliation(s)
- Thanh N Dang
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, 70808, USA
| | - Rafael P Tiongco
- Tulane University School of Medicine, New Orleans, Louisiana, 70118, USA
| | - Loren M Brown
- Department of Surgery, Louisiana State University Health Science Center, New Orleans, Louisiana, 70112, USA
| | - Jessica L Taylor
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, 70808, USA
| | - John M Lyons
- Our Lady of the Lake Medical Center, Baton Rouge, Louisiana, 70808, USA
| | - Frank H Lau
- Department of Surgery, Louisiana State University Health Science Center, New Orleans, Louisiana, 70112, USA.
| | - Z Elizabeth Floyd
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, 70808, USA.
| |
Collapse
|
49
|
Abstract
Obesity has reached epidemic proportions and is a major contributor to insulin resistance (IR) and type 2 diabetes (T2D). Importantly, IR and T2D substantially increase the risk of cardiovascular (CV) disease. Although there are successful approaches to maintain glycemic control, there continue to be increased CV morbidity and mortality associated with metabolic disease. Therefore, there is an urgent need to understand the cellular and molecular processes that underlie cardiometabolic changes that occur during obesity so that optimal medical therapies can be designed to attenuate or prevent the sequelae of this disease. The vascular endothelium is in constant contact with the circulating milieu; thus, it is not surprising that obesity-driven elevations in lipids, glucose, and proinflammatory mediators induce endothelial dysfunction, vascular inflammation, and vascular remodeling in all segments of the vasculature. As cardiometabolic disease progresses, so do pathological changes in the entire vascular network, which can feed forward to exacerbate disease progression. Recent cellular and molecular data have implicated the vasculature as an initiating and instigating factor in the development of several cardiometabolic diseases. This Review discusses these findings in the context of atherosclerosis, IR and T2D, and heart failure with preserved ejection fraction. In addition, novel strategies to therapeutically target the vasculature to lessen cardiometabolic disease burden are introduced.
Collapse
|
50
|
Rabhi N, Desevin K, Belkina AC, Tilston-Lunel A, Varelas X, Layne MD, Farmer SR. Obesity-induced senescent macrophages activate a fibrotic transcriptional program in adipocyte progenitors. Life Sci Alliance 2022; 5:5/5/e202101286. [PMID: 35181634 PMCID: PMC8860101 DOI: 10.26508/lsa.202101286] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
This study demonstrates that senescent CD9+ macrophages in obese visceral fat of mice secrete osteopontin that promotes ECM deposition by adipogenic progenitor cells expressing Pdgfra and Pdgfrb. Adipose tissue fibrosis is regulated by the chronic and progressive metabolic imbalance caused by differences in caloric intake and energy expenditure. By exploring the cellular heterogeneity within fibrotic adipose tissue, we demonstrate that early adipocyte progenitor cells expressing both platelet-derived growth factor receptor (PDGFR) α and β are the major contributors to extracellular matrix deposition. We show that the fibrotic program is promoted by senescent macrophages. These macrophages were enriched in the fibrotic stroma and exhibit a distinct expression profile. Furthermore, we demonstrate that these cells display a blunted phagocytotic capacity and acquire a senescence-associated secretory phenotype. Finally, we determined that osteopontin, which was expressed by senescent macrophages in the fibrotic environment promoted progenitor cell proliferation, fibrotic gene expression, and inhibited adipogenesis. Our work reveals that obesity promotes macrophage senescence and provides a conceptual framework for the discovery of rational therapeutic targets for metabolic and inflammatory disease associated with obesity.
Collapse
Affiliation(s)
- Nabil Rabhi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Kathleen Desevin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Anna C Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Matthew D Layne
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Stephen R Farmer
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|