1
|
Zhong Y, Yan J, Lei Y, Zhang R, Abudurexiti A, Qi S, Hou W, Ma X. Lactucin and lactucopicrin ameliorate obesity in high-fat diet fed mice by promoting white adipose tissue browning through the activation of the AMPK/SIRT1/PGC-1α pathway. J Nutr Biochem 2025; 139:109851. [PMID: 39909319 DOI: 10.1016/j.jnutbio.2025.109851] [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: 06/20/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
Lactucin and lactucopicrin are the characteristic lipid-lowering active components found in Cichorium glandulosum. However, their effects and underlying mechanisms in obesity remain unclear. In the present study, C57BL/6J mice were simultaneously subjected to a high-fat diet (HFD) and treated with drugs to investigate the impacts of lactucin and lactucopicrin on HFD-induced obese mice. The results demonstrated that in HFD obese mice, lactucin and lactucopicrin significantly decreased body weight and the weights of adipose tissues, improved serum metabolic parameters, and increased the content of irisin. Regarding the intermediate metabolites of intestinal flora, which are closely associated with white adipose tissue (WAT) browning, lactucin and lactucopicrin treatment led to a reduction in the levels of 12-α-OH/non-12-α-OH bile acids (BAs) and also tended to enhance the levels of short-chain fatty acids (SCFAs). qRT-PCR results indicated that lactucin and lactucopicrin treatment elevated the expression levels of genes related to beige fat markers, thermogenesis, mitochondrial biogenesis, and lipolysis in WAT, as well as those of thermogenesis and lipolysis genes in brown adipose tissue (BAT). Western blot analysis revealed that lactucin and lactucopicrin up-regulated the expression of uncoupling protein 1 (UCP1), the core protein in thermogenesis, in both WAT and BAT. Moreover, they also up-regulated the expression levels of AMP-activated kinase (AMPK), sirtuin 1 (SIRT1), and PPARγ coactivator 1-alpha (PGC-1α), which are key pathway proteins involved in WAT browning. Furthermore, 16S rRNA sequencing results showed that in HFD obese mice, lactucin and lactucopicrin improved the composition and function of the intestinal microbiota. In conclusion, lactucin and lactucopicrin may promote WAT browning by activating the AMPK/SIRT1/PGC-1α pathway, thereby ameliorating obesity in HFD mice.
Collapse
Affiliation(s)
- Yewei Zhong
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Junlin Yan
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Yi Lei
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Rui Zhang
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | | | - Shuwen Qi
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Wenhui Hou
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Xiaoli Ma
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
2
|
Sleiman L, Dinescu S. Role of Non-Coding RNAs in White and Brown Adipose Tissue Differentiation and Development. Noncoding RNA 2025; 11:30. [PMID: 40407588 PMCID: PMC12101253 DOI: 10.3390/ncrna11030030] [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: 02/28/2025] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Adipocyte differentiation is a complex process in which pluripotent mesenchymal stem cells (MSCs) differentiate and develop into mature fat cells, also known as adipocytes. This process is controlled by various transcription factors, hormones, and signaling molecules that regulate the development of these cells. Recently, an increasing number of non-coding RNAs (ncRNAs), especially microRNAs (miRNAs), have been established to be involved in the regulation of many biological processes, including adipocyte differentiation, development, metabolism, and energy homeostasis of white and brown adipose tissue. Several in vitro and in vivo studies reported the significant role of ncRNAs in either promoting or inhibiting adipocyte differentiation into white or brown fat cells by targeting specific transcription factors and regulating the expression of key adipogenic genes. Identifying the function of ncRNAs and their subsequent targets contributes to our understanding of how these molecules can be used as potential biomarkers and tools for therapies against obesity, diabetes, and other diseases related to obesity. This could also contribute to advancements in tissue-engineering based treatments. In this review, we intended to present an up-to-date comprehensive literature overview of the role of ncRNAs, including miRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), focusing particularly on miRNAs, in regulating the differentiation and development of cells into white and brown adipose tissue. In addition, we further discuss the potential use of these molecules as biomarkers for the development of novel therapeutic strategies for future personalized treatment options for patients.
Collapse
Affiliation(s)
- Lea Sleiman
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
- Research Institute of the University of Bucharest (ICUB), 050663 Bucharest, Romania
| |
Collapse
|
3
|
Rivera-Alvarez I, Vázquez-Lizárraga R, Mendoza-Viveros L, Sotelo-Rivera I, Viveros-Ruiz TL, Morales-Maza J, Orozco L, Romano MC, Noriega LG, Tovar AR, Aguilar-Arnal L, Cruz-Bautista I, Aguilar-Salinas C, Orozco-Solis R. Transcriptional dynamics in type 2 diabetes progression is linked with circadian, thermogenic, and cellular stress in human adipose tissue. Commun Biol 2025; 8:398. [PMID: 40057615 PMCID: PMC11890630 DOI: 10.1038/s42003-025-07709-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/10/2025] [Indexed: 04/03/2025] Open
Abstract
The prevalence of type 2 diabetes (T2D) has increased significantly over the past three decades, with an estimated 30-40% of cases remaining undiagnosed. Brown and beige adipose tissues are known for their remarkable catabolic capacity, and their ability to diminish blood glucose plasma concentration. Beige adipose tissue can be differentiated from adipose-derived stem cells or through transdifferentiation from white adipocytes. However, the impact of T2D progression on beige adipocytes' functional capacity remains unclear. Transcriptomic profiling of subcutaneous adipose tissue biopsies from healthy normal-weight, obese, prediabetic obese, and obese subjects diagnosed with T2D, reveals a progressive alteration in cellular processes associated with catabolic metabolism, circadian rhythms, thermogenesis-related signaling pathways, cellular stress, and inflammation. MAX is a potential transcription factor that links inflammation with the circadian clock and thermogenesis during the progression of T2D. This study unveils an unrecognized transcriptional circuit that increasingly disrupts subcutaneous adipose tissue oxidative capacity during the progression of T2D. These findings could open new research venues for developing chrono-pharmaceutical strategies to treat and prevent T2D.
Collapse
Affiliation(s)
| | - Rosa Vázquez-Lizárraga
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| | - Lucía Mendoza-Viveros
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosi, S.L.P., México
| | | | - Tannia L Viveros-Ruiz
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Jesús Morales-Maza
- Departamento de Cirugía Endocrina, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
| | - Marta C Romano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y Estudios Avanzados (CINVESTAV), México City, México
| | - Lilia G Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Lorena Aguilar-Arnal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), México City, México
| | - Ivette Cruz-Bautista
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
| | - Carlos Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), México City, México
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, México City, México
| | - Ricardo Orozco-Solis
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México.
- Centro de Investigación sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), México City, México.
| |
Collapse
|
4
|
Pani S, Senapati U, Sahu B, Pati B, Swalsingh G, Pani P, Bindhani BK, Achary KG, Bal NC. Developmental overlap between skeletal muscle maturation and perirenal fat brown-to-white transition in goats: Exploring the role of Myf-5. Biochimie 2025; 228:1-7. [PMID: 39121901 DOI: 10.1016/j.biochi.2024.08.005] [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: 04/24/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
In mammals, skeletal muscles (SkMs) and adipose tissues regulate energy homeostasis and share developmental origins. Notably, the perirenal adipose tissue (PRAT) depot has been reported to display adipocyte heterogeneity: while some originated from Myogenic factor 5 (Myf-5) expressing progenitors, others did not. Our study examines the expression and distribution of Myf-5 using immunohistochemical staining and western blotting of PRAT, gastrocnemius, and trapezius from goat at various developmental stages. Contrary to earlier beliefs, functionally divergent SkM gastrocnemius and trapezius showed similar Myf-5 expressional pattern. SkM abundantly expresses Myf-5 in developing myocytes which gradually becomes limited to the nucleus of myogenic stem cells and is retained only in a few differentiated postnatal fibers. During the same period, PRAT displays a unique brown-to-white transition. PRAT exhibited an elevated expression of Myf-5 during prenatal periods, which declines thereafter and becomes negligible during adulthood where it gets fully enriched white adipocytes. The reduction of Myf-5 during the neonatal period was common to all three tissues. However, Myf-5 expression was retained in some of the differentiated myofibers while it was undetectable in adult PRAT. These observations suggest a possible developmental interplay between adipose tissue and SkM where Myf-5 might be a major regulator.
Collapse
Affiliation(s)
- Sunil Pani
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | - Unmod Senapati
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | - Bijayashree Sahu
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | - Benudhara Pati
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | | | - Punyadhara Pani
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | | | | | - Naresh C Bal
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India.
| |
Collapse
|
5
|
Malicka A, Ali A, MacCannell ADV, Roberts LD. Brown and beige adipose tissue-derived metabokine and lipokine inter-organ signalling in health and disease. Exp Physiol 2024. [PMID: 39591977 DOI: 10.1113/ep092008] [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: 07/19/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024]
Abstract
Adipose tissue has an established endocrine function through the secretion of adipokines. However, a role for bioactive metabolites and lipids, termed metabokines and lipokines, is emerging in adipose tissue-mediated autocrine, paracrine and endocrine signalling and inter-organ communication. Traditionally seen as passive entities, metabolites are now recognized for their active roles in regulating cellular signalling and local and systemic metabolism. Distinct from white adipose tissue, specific endocrine functions have been attributed to thermogenic brown and beige adipose tissues. Brown and beige adipose tissues have been identified as sources of metabokines and lipokines, which influence diverse metabolic pathways, such as fatty acid β-oxidation, mitochondrial function and glucose homeostasis, across a range of tissues, including skeletal muscle, adipose tissue and heart. This review explores the intricate signalling mechanisms of brown and beige adipose tissue-derived metabokines and lipokines, emphasizing their roles in maintaining metabolic homeostasis and their potential dysregulation in metabolic diseases. Furthermore, we discuss the therapeutic potential of targeting these pathways, proposing that precise modulation of metabokine receptors and transporters could offer superior specificity and efficacy in comparison to conventional approaches, such as β-adrenergic signalling-stimulated activation of brown adipose tissue thermogenesis. Understanding the complex interactions between adipokines, metabokines and lipokines is essential for developing a systems-level approach to new interventions for metabolic disorders, underscoring the need for continued research in this rapidly evolving field.
Collapse
Affiliation(s)
- Anna Malicka
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Aysha Ali
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
Navarro-Perez J, Carobbio S. Adipose tissue-derived stem cells, in vivo and in vitro models for metabolic diseases. Biochem Pharmacol 2024; 222:116108. [PMID: 38438053 DOI: 10.1016/j.bcp.2024.116108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
The primary role of adipose tissue stem cells (ADSCs) is to support the function and homeostasis of adipose tissue in physiological and pathophysiological conditions. However, when ADSCs become dysfunctional in diseases such as obesity and cancer, they become impaired, undergo signalling changes, and their epigenome is altered, which can have a dramatic effect on human health. In more recent years, the therapeutic potential of ADSCs in regenerative medicine, wound healing, and for treating conditions such as cancer and metabolic diseases has been extensively investigated with very promising results. ADSCs have also been used to generate two-dimensional (2D) and three-dimensional (3D) cellular and in vivo models to study adipose tissue biology and function as well as intracellular communication. Characterising the biology and function of ADSCs, how it is altered in health and disease, and its therapeutic potential and uses in cellular models is key for designing intervention strategies for complex metabolic diseases and cancer.
Collapse
|
10
|
Bahadoran Z, Mirmiran P, Ghasemi A. Adipose organ dysfunction and type 2 diabetes: Role of nitric oxide. Biochem Pharmacol 2024; 221:116043. [PMID: 38325496 DOI: 10.1016/j.bcp.2024.116043] [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: 10/30/2023] [Revised: 01/07/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Adipose organ, historically known as specialized lipid-handling tissue serving as the long-term fat depot, is now appreciated as the largest endocrine organ composed of two main compartments, i.e., subcutaneous and visceral adipose tissue (AT), madding up white and beige/brown adipocytes. Adipose organ dysfunction manifested as maldistribution of the compartments, hypertrophic, hypoxic, inflamed, and insulin-resistant AT, contributes to the development of type 2 diabetes (T2D). Here, we highlight the role of nitric oxide (NO·) in AT (dys)function in relation to developing T2D. The key aspects determining lipid and glucose homeostasis in AT depend on the physiological levels of the NO· produced via endothelial NO· synthases (eNOS). In addition to decreased NO· bioavailability (via decreased expression/activity of eNOS or scavenging NO·), excessive NO· produced by inducible NOS (iNOS) in response to hypoxia and AT inflammation may be a critical interfering factor diverting NO· signaling to the formation of reactive oxygen and nitrogen species, resulting in AT and whole-body metabolic dysfunction. Pharmacological approaches boosting AT-NO· availability at physiological levels (by increasing NO· production and its stability), as well as suppression of iNOS-NO· synthesis, are potential candidates for developing NO·-based therapeutics in T2D.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Proença C, Freitas M, Rocha S, Ferreira de Oliveira JMP, Carvalho F, Fernandes E. Unravelling the Influence of Endocrine-Disrupting Chemicals on Obesity Pathophysiology Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:883-918. [PMID: 39287876 DOI: 10.1007/978-3-031-63657-8_30] [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
Obesity represents a global health concern, affecting individuals of all age groups across the world. The prevalence of excess weight and obesity has escalated to pandemic proportions, leading to a substantial increase in the incidence of various comorbidities, such as cardiovascular diseases, type 2 diabetes, and cancer. This chapter seeks to provide a comprehensive exploration of the pathways through which endocrine-disrupting chemicals can influence the pathophysiology of obesity. These mechanisms encompass aspects such as the regulation of food intake and appetite, intestinal fat absorption, lipid metabolism, and the modulation of inflammation. This knowledge may help to elucidate the role of exogenous molecules in both the aetiology and progression of obesity.
Collapse
Affiliation(s)
- Carina Proença
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Sílvia Rocha
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - José Miguel P Ferreira de Oliveira
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| |
Collapse
|
12
|
Huang Z, Gu C, Zhang Z, Arianti R, Swaminathan A, Tran K, Battist A, Kristóf E, Ruan HB. Supraclavicular brown adipocytes originate from Tbx1+ myoprogenitors. PLoS Biol 2023; 21:e3002413. [PMID: 38048357 PMCID: PMC10721186 DOI: 10.1371/journal.pbio.3002413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/14/2023] [Accepted: 10/31/2023] [Indexed: 12/06/2023] Open
Abstract
Brown adipose tissue (BAT) dissipates energy as heat, contributing to temperature control, energy expenditure, and systemic homeostasis. In adult humans, BAT mainly exists in supraclavicular areas and its prevalence is associated with cardiometabolic health. However, the developmental origin of supraclavicular BAT remains unknown. Here, using genetic cell marking in mice, we demonstrate that supraclavicular brown adipocytes do not develop from the Pax3+/Myf5+ epaxial dermomyotome that gives rise to interscapular BAT (iBAT). Instead, the Tbx1+ lineage that specifies the pharyngeal mesoderm marks the majority of supraclavicular brown adipocytes. Tbx1Cre-mediated ablation of peroxisome proliferator-activated receptor gamma (PPARγ) or PR/SET Domain 16 (PRDM16), components of the transcriptional complex for brown fat determination, leads to supraclavicular BAT paucity or dysfunction, thus rendering mice more sensitive to cold exposure. Moreover, human deep neck BAT expresses higher levels of the TBX1 gene than subcutaneous neck white adipocytes. Taken together, our observations reveal location-specific developmental origins of BAT depots and call attention to Tbx1+ lineage cells when investigating human relevant supraclavicular BAT.
Collapse
Affiliation(s)
- Zan Huang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Chenxin Gu
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Zengdi Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Rini Arianti
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Aneesh Swaminathan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Kevin Tran
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Alex Battist
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Endre Kristóf
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| |
Collapse
|
13
|
You W, Xu Z, Chen W, Yang X, Liu S, Wang L, Tu Y, Zhou Y, Valencak TG, Wang Y, Kuang S, Shan T. Cellular and Transcriptional Dynamics during Brown Adipose Tissue Regeneration under Acute Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0268. [PMID: 38434240 PMCID: PMC10907023 DOI: 10.34133/research.0268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/18/2023] [Indexed: 03/05/2024]
Abstract
Brown adipose tissue (BAT) is the major site of non-shivering thermogenesis and crucial for systemic metabolism. Under chronic cold exposures and high-fat diet challenges, BAT undergoes robust remodeling to adapt to physiological demands. However, whether and how BAT regenerates after acute injuries are poorly understood. Here, we established a novel BAT injury and regeneration model (BAT-IR) in mice and performed single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq to determine cellular and transcriptomic dynamics during BAT-IR. We further defined distinct fibro-adipogenic and myeloid progenitor populations contributing to BAT regeneration. Cell trajectory and gene expression analyses uncovered the involvement of MAPK, Wnt, and Hedgehog (Hh) signaling pathways in BAT regeneration. We confirmed the role of Hh signaling in BAT development through Myf5Cre-mediated conditional knockout (cKO) of the Sufu gene to activate Hh signaling in BAT and muscle progenitors. Our BAT-IR model therefore provides a paradigm to identify conserved cellular and molecular mechanisms underlying BAT development and remodeling.
Collapse
Affiliation(s)
- Wenjing You
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Ziye Xu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- Department of Laboratory Medicine, the First Affiliated Hospital, College of Medicine,
Zhejiang University, Hangzhou, China
| | - Wentao Chen
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Xin Yang
- Department of Animal Sciences,
Purdue University, West Lafayette, IN, USA
| | - Shiqi Liu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yuang Tu
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yanbing Zhou
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | | | - Yizhen Wang
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Shihuan Kuang
- Department of Animal Sciences,
Purdue University, West Lafayette, IN, USA
| | - Tizhong Shan
- College of Animal Sciences,
Zhejiang University, Hangzhou, China
- The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| |
Collapse
|
14
|
Nie T, Lu J, Zhang H, Mao L. Latest advances in the regulatory genes of adipocyte thermogenesis. Front Endocrinol (Lausanne) 2023; 14:1250487. [PMID: 37680891 PMCID: PMC10482227 DOI: 10.3389/fendo.2023.1250487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
An energy imbalance cause obesity: more energy intake or less energy expenditure, or both. Obesity could be the origin of many metabolic disorders, such as type 2 diabetes and cardiovascular disease. UCP1 (uncoupling protein1), which is highly and exclusively expressed in the thermogenic adipocytes, including beige and brown adipocytes, can dissipate proton motive force into heat without producing ATP to increase energy expenditure. It is an attractive strategy to combat obesity and its related metabolic disorders by increasing non-shivering adipocyte thermogenesis. Adipocyte thermogenesis has recently been reported to be regulated by several new genes. This work provided novel and potential targets to activate adipocyte thermogenesis and resist obesity, such as secreted proteins ADISSP and EMC10, enzyme SSU72, etc. In this review, we have summarized the latest research on adipocyte thermogenesis regulation to shed more light on this topic.
Collapse
Affiliation(s)
- Tao Nie
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Jinli Lu
- Scientific Research Center, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hua Zhang
- Department of Medical Iconography, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Liufeng Mao
- Scientific Research Center, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
15
|
Lin YC, Hou YC, Wang HC, Shan YS. New insights into the role of adipocytes in pancreatic cancer progression: paving the way towards novel therapeutic targets. Theranostics 2023; 13:3925-3942. [PMID: 37554282 PMCID: PMC10405844 DOI: 10.7150/thno.82911] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/21/2023] [Indexed: 08/10/2023] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal malignancies across the world, which is due to delayed diagnosis and resistance to current therapies. The interactions between pancreatic tumor cells and their tumor microenvironment (TME) allow cancer cells to escape from anti-cancer therapies, leading to difficulties in treating PC. With endocrine function and lipid storage capacity, adipose tissue can maintain energy homeostasis. Direct or indirect interaction between adipocytes and PC cells leads to adipocyte dysfunction characterized by morphological change, fat loss, abnormal adipokine secretion, and fibroblast-like transformation. Various adipokines released from dysfunctional adipocytes have been reported to promote proliferation, invasion, metastasis, stemness, and chemoresistance of PC cells via different mechanisms. Additional lipid outflow from adipocytes can be taken into the TME and thus alter the metabolism in PC cells and surrounding stromal cells. Besides, the trans-differentiation potential enables adipocytes to turn into various cell types, which may give rise to an inflammatory response as well as extracellular matrix reorganization to modulate tumor burden. Understanding the molecular basis behind the protumor functions of adipocytes in PC may offer new therapeutic targets.
Collapse
Affiliation(s)
- Yu-Chun Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Hao-Chen Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Medical Imaging Center, Innovation Headquarter, National Cheng Kung University; Tainan 704, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| |
Collapse
|
16
|
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
|
17
|
Grillo E, Ravelli C, Colleluori G, D'Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev 2023; 69:51-60. [PMID: 36155165 DOI: 10.1016/j.cytogfr.2022.09.004] [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: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Francesco D'Agostino
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Domenichini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
18
|
Hoang AC, Sasi-Szabó L, Pál T, Szabó T, Diedrich V, Herwig A, Landgraf K, Körner A, Röszer T. Mitochondrial RNA stimulates beige adipocyte development in young mice. Nat Metab 2022; 4:1684-1696. [PMID: 36443525 PMCID: PMC9771821 DOI: 10.1038/s42255-022-00683-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 10/10/2022] [Indexed: 11/30/2022]
Abstract
Childhood obesity is a serious public health crisis and a critical factor that determines future obesity prevalence. Signals affecting adipocyte development in early postnatal life have a strong potential to trigger childhood obesity; however, these signals are still poorly understood. We show here that mitochondrial (mt)RNA efflux stimulates transcription of nuclear-encoded genes for mitobiogenesis and thermogenesis in adipocytes of young mice and human infants. While cytosolic mtRNA is a potential trigger of the interferon (IFN) response, young adipocytes lack such a response to cytosolic mtRNA due to the suppression of IFN regulatory factor (IRF)7 expression by vitamin D receptor signalling. Adult and obese adipocytes, however, strongly express IRF7 and mount an IFN response to cytosolic mtRNA. In turn, suppressing IRF7 expression in adult adipocytes restores mtRNA-induced mitobiogenesis and thermogenesis and eventually mitigates obesity. Retrograde mitochondrion-to-nucleus signalling by mtRNA is thus a mechanism to evoke thermogenic potential during early adipocyte development and to protect against obesity.
Collapse
Affiliation(s)
| | - László Sasi-Szabó
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor Pál
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Szabó
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Annika Herwig
- Institute of Neurobiology, Ulm University, Ulm, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Tamás Röszer
- Institute of Neurobiology, Ulm University, Ulm, Germany.
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
19
|
Sreekumar S, Vijayan V, Singh F, Sudhakar M, Lakra R, Korrapati PS, Kiran MS. White to brown adipocyte transition mediated by Apigenin via VEGF-PRDM16 signaling. J Cell Biochem 2022; 123:1793-1807. [PMID: 35926149 DOI: 10.1002/jcb.30316] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/08/2022]
Abstract
The dysregulated energy metabolism in white adipose tissues results in derangement of biological signaling resulting in obesity. Lack of vascularization in these white adipose tissues is one of the major reasons for dysregulated energy metabolism. Not much work has been done in this direction to understand the role of angiogenesis in white adipose tissue metabolism. In the present study, we evaluated the effect of angiogenic modulator in the metabolism of white adipocyte (WAC). Bioactive Apigenin was selected and its angiogenic ability was studied. Apigenin was shown to be highly proangiogenic hence the effect of Apigenin on de novo and trans-differentiation of WAT was studied. Apigenin showed enhanced de novo differentiation and trans-differentiation of mouse WAC into brown-like phenotype. To understand the effect of Apigenin on adipose tissue vasculature, coculture studies were conducted. Cross talk between endothelial cell and adipocytes were observed in coculture studies. Gene expression studies of cocultured cells revealed that browning of WAC occurred by triggering the expression of Vascular endothelial growth factor A. The study provides a new insight for inducing metabolic shift in WACs by modulation of angiogenesis in WAC microenvironment by the upregulation of PRDM16 cascade to trigger browning for the treatment of obesity.
Collapse
Affiliation(s)
- Sreelekshmi Sreekumar
- Biological Materials Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, Tamil Nadu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vinu Vijayan
- Biological Materials Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, Tamil Nadu, India
| | - Fathe Singh
- Biological Materials Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, Tamil Nadu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manu Sudhakar
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (DU), Chennai, India
| | - Rachita Lakra
- Biological Materials Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, Tamil Nadu, India
| | - Purna Sai Korrapati
- Biological Materials Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, Tamil Nadu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manikantan Syamala Kiran
- Biological Materials Laboratory, Council of Scientific and Industrial Research-Central Leather Research Institute, Chennai, Tamil Nadu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
20
|
Factors Associated with White Fat Browning: New Regulators of Lipid Metabolism. Int J Mol Sci 2022; 23:ijms23147641. [PMID: 35886989 PMCID: PMC9325132 DOI: 10.3390/ijms23147641] [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: 06/07/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
Mammalian adipose tissue can be divided into white and brown adipose tissue based on its colour, location, and cellular structure. Certain conditions, such as sympathetic nerve excitement, can induce the white adipose adipocytes into a new type of adipocytes, known as beige adipocytes. The process, leading to the conversion of white adipocytes into beige adipocytes, is called white fat browning. The dynamic balance between white and beige adipocytes is closely related to the body’s metabolic homeostasis. Studying the signal transduction pathways of the white fat browning might provide novel ideas for the treatment of obesity and alleviation of obesity-related glucose and lipid metabolism disorders. This article aimed to provide an overview of recent advances in understanding white fat browning and the role of BAT in lipid metabolism.
Collapse
|
21
|
Fung CW, Zhou S, Zhu H, Wei X, Wu Z, Wu AR. Cell fate determining molecular switches and signaling pathways in Pax7-expressing somitic mesoderm. Cell Discov 2022; 8:61. [PMID: 35764624 PMCID: PMC9240041 DOI: 10.1038/s41421-022-00407-0] [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: 02/09/2021] [Accepted: 03/28/2022] [Indexed: 11/09/2022] Open
Abstract
During development, different cell types originate from a common progenitor at well-defined time points. Previous lineage-tracing of Pax7+ progenitors from the somitic mesoderm has established its developmental trajectory towards the dermis, brown adipocytes, and skeletal muscle in the dorsal trunk; yet the molecular switches and mechanisms guiding the differentiation into different lineages remain unknown. We performed lineage-tracing of Pax7-expressing cells in mouse embryos at E9.5 and profiled the transcriptomes of Pax7-progenies on E12.5, E14.5, and E16.5 at single-cell level. Analysis of single-cell transcriptomic data at multiple time points showed temporal-specific differentiation events toward muscle, dermis, and brown adipocyte, identified marker genes for putative progenitors and revealed transcription factors that could drive lineage-specific differentiation. We then utilized a combination of surface markers identified in the single-cell data, Pdgfra, Thy1, and Cd36, to enrich brown adipocytes, dermal fibroblasts, and progenitors specific for these two cell types at E14.5 and E16.5. These enriched cell populations were then used for further culture and functional assays in vitro, in which Wnt5a and Rgcc are shown to be important factors that could alter lineage decisions during embryogenesis. Notably, we found a bipotent progenitor population at E14.5, having lineage potentials towards both dermal fibroblasts and brown adipocytes. They were termed eFAPs (embryonic fibro/adipogenic progenitors) as they functionally resemble adult fibro/adipogenic progenitors. Overall, this study provides further understanding of the Pax7 lineage during embryonic development using a combination of lineage tracing with temporally sampled single-cell transcriptomics.
Collapse
|
22
|
Altınova AE. Beige Adipocyte as the Flame of White Adipose Tissue: Regulation of Browning and Impact of Obesity. J Clin Endocrinol Metab 2022; 107:e1778-e1788. [PMID: 34967396 DOI: 10.1210/clinem/dgab921] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Indexed: 11/19/2022]
Abstract
Beige adipocyte, the third and relatively new type of adipocyte, can emerge in white adipose tissue (WAT) under thermogenic stimulations that is termed as browning of WAT. Recent studies suggest that browning of WAT deserves more attention and therapies targeting browning of WAT can be helpful for reducing obesity. Beyond the major inducers of browning, namely cold and β 3-adrenergic stimulation, beige adipocytes are affected by several factors, and excess adiposity per se may also influence the browning process. The objective of the present review is to provide an overview of recent clinical and preclinical studies on the hormonal and nonhormonal factors that affect the browning of WAT. This review further focuses on the role of obesity per se on browning process.
Collapse
Affiliation(s)
- Alev Eroğlu Altınova
- Gazi University Faculty of Medicine, Department of Endocrinology and Metabolism, 06500 Ankara, Turkey
| |
Collapse
|
23
|
Colleluori G, Perugini J, Giordano A, Cinti S. From Obesity to Diabetes: The Role of the Adipose Organ. Handb Exp Pharmacol 2022; 274:75-92. [PMID: 35044536 DOI: 10.1007/164_2021_572] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity is a complex, multifactorial, and relapsing disease whose prevalence has tripled during the last decades and whose incidence is expected to further increase. For these reasons, obesity is considered as a real pandemic, deeply burdening the global health-care systems. From a pathophysiological standpoint obesity is the result of a chronic-positive energy balance which in turn leads to an excessive accumulation of lipids, not only within the adipose organ, but also in different cytotypes, a phenomenon leading to lipotoxicity that deeply compromises several cellular and organs functions. Obesity is therefore associated with over 200 medical complications, including insulin resistance and type 2 diabetes mellitus (T2DM) and represents the fifth leading cause of death worldwide. In this review, we describe the main pathophysiological mechanisms linking obesity-induced adipose organ dysfunction to insulin resistance and T2DM.
Collapse
Affiliation(s)
- Georgia Colleluori
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Ancona, Italy.
| |
Collapse
|
24
|
Li F, Jing J, Movahed M, Cui X, Cao Q, Wu R, Chen Z, Yu L, Pan Y, Shi H, Shi H, Xue B. Epigenetic interaction between UTX and DNMT1 regulates diet-induced myogenic remodeling in brown fat. Nat Commun 2021; 12:6838. [PMID: 34824202 PMCID: PMC8617140 DOI: 10.1038/s41467-021-27141-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/05/2021] [Indexed: 02/04/2023] Open
Abstract
Brown adipocytes share the same developmental origin with skeletal muscle. Here we find that a brown adipocyte-to-myocyte remodeling also exists in mature brown adipocytes, and is induced by prolonged high fat diet (HFD) feeding, leading to brown fat dysfunction. This process is regulated by the interaction of epigenetic pathways involving histone and DNA methylation. In mature brown adipocytes, the histone demethylase UTX maintains persistent demethylation of the repressive mark H3K27me3 at Prdm16 promoter, leading to high Prdm16 expression. PRDM16 then recruits DNA methyltransferase DNMT1 to Myod1 promoter, causing Myod1 promoter hypermethylation and suppressing its expression. The interaction between PRDM16 and DNMT1 coordinately serves to maintain brown adipocyte identity while repressing myogenic remodeling in mature brown adipocytes, thus promoting their active brown adipocyte thermogenic function. Suppressing this interaction by HFD feeding induces brown adipocyte-to-myocyte remodeling, which limits brown adipocyte thermogenic capacity and compromises diet-induced thermogenesis, leading to the development of obesity.
Collapse
Affiliation(s)
- Fenfen Li
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Jia Jing
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Miranda Movahed
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Xin Cui
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Qiang Cao
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Rui Wu
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Ziyue Chen
- grid.256304.60000 0004 1936 7400Department of Computer Science, Georgia State University, Atlanta, GA 30303 USA
| | - Liqing Yu
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Yi Pan
- grid.256304.60000 0004 1936 7400Department of Computer Science, Georgia State University, Atlanta, GA 30303 USA ,grid.458489.c0000 0001 0483 7922Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 P.R. China
| | - Huidong Shi
- grid.410427.40000 0001 2284 9329Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA ,grid.410427.40000 0001 2284 9329Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912 USA
| | - Hang Shi
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| | - Bingzhong Xue
- grid.256304.60000 0004 1936 7400Department of Biology, Georgia State University, Atlanta, GA 30303 USA
| |
Collapse
|
25
|
Wang X, Cai H, Shui S, Lin Y, Wang F, Wang L, Chen J, Liu J. Chrysin Stimulates Subcutaneous Fat Thermogenesis in Mice by Regulating PDGFRα and MicroRNA Expressions. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5897-5906. [PMID: 34027663 DOI: 10.1021/acs.jafc.1c01130] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The activation of adipose tissue browning and thermogenesis provides a new strategy to counter obesity and associated metabolic diseases. Here, a natural flavonoid chrysin is used as the supplement of a high-fat diet (HFD). Dietary chrysin alleviates adiposity and insulin resistance in HFD-fed mice. Meanwhile, dietary chrysin elevates systemic energy expenditure and enhances the uncoupling protein-1 (UCP1) level in subcutaneous adipose tissue (SAT), which is accompanied by the increased thermogenic program, beige preadipocyte number, and angiogenesis in SAT. Dietary chrysin also induces the expression of SAT platelet-derived growth factor receptor α (PDGFRα), which commits adipose progenitor cells to differentiate into beige or white adipocytes in response to various environmental signals. Double immunofluorescent staining for UCP1 and PDGFRα reveals that chrysin elevates the number of UCP1+PDGFRα+ beige progenitors in SAT. Further, chrysin treatment reverses the effects of the specific PDGFRα inhibitor imatinib on browning differentiation of stromal vascular fraction cells from SAT. Finally, chrysin-induced adipocyte browning is correlated with the expressions of microRNAs as PDGFRα inhibitors or thermogenesis suppressors. In conclusion, dietary chrysin promotes subcutaneous adipocyte browning and systematic energy expenditure by regulating PDGFRα and microRNA expressions in HFD-fed mice.
Collapse
Affiliation(s)
- Xin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Hao Cai
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Shanshan Shui
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Yan Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Fangbin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Lu Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230009, China
- Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei 230009, China
| |
Collapse
|
26
|
Gui C, Parson J, Meyer GA. Harnessing adipose stem cell diversity in regenerative medicine. APL Bioeng 2021; 5:021501. [PMID: 33834153 PMCID: PMC8018797 DOI: 10.1063/5.0038101] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/26/2021] [Indexed: 12/20/2022] Open
Abstract
Since the first isolation of mesenchymal stem cells from lipoaspirate in the early 2000s, adipose tissue has been a darling of regenerative medicine. It is abundant, easy to access, and contains high concentrations of stem cells (ADSCs) exhibiting multipotency, proregenerative paracrine signaling, and immunomodulation-a winning combination for stem cell-based therapeutics. While basic science, preclinical and clinical findings back up the translational potential of ADSCs, the vast majority of these used cells from a single location-subcutaneous abdominal fat. New data highlight incredible diversity in the adipose morphology and function in different anatomical locations or depots. Even in isolation, ADSCs retain a memory of this diversity, suggesting that the optimal adipose source material for ADSC isolation may be application specific. This review discusses our current understanding of the heterogeneity in the adipose organ, how that heterogeneity translates into depot-specific ADSC characteristics, and how atypical ADSC populations might be harnessed for regenerative medicine applications. While our understanding of the breadth of ADSC heterogeneity is still in its infancy, clear trends are emerging for application-specific sourcing to improve regenerative outcomes.
Collapse
Affiliation(s)
- Chang Gui
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63110, USA
| | - Jacob Parson
- Program in Physical Therapy, Washington University in St. Louis, St. Louis, Missouri 63110, USA
| | - Gretchen A. Meyer
- Author to whom correspondence should be addressed:. Tel.: (314) 286-1425. Fax: (314) 747-0674
| |
Collapse
|
27
|
Brandão BB, Poojari A, Rabiee A. Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential. Int J Mol Sci 2021; 22:5906. [PMID: 34072788 PMCID: PMC8198523 DOI: 10.3390/ijms22115906] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The concerning worldwide increase of obesity and chronic metabolic diseases, such as T2D, dyslipidemia, and cardiovascular disease, motivates further investigations into preventive and alternative therapeutic approaches. Over the past decade, there has been growing evidence that the formation and activation of thermogenic adipocytes (brown and beige) may serve as therapy to treat obesity and its associated diseases owing to its capacity to increase energy expenditure and to modulate circulating lipids and glucose levels. Thus, understanding the molecular mechanism of brown and beige adipocytes formation and activation will facilitate the development of strategies to combat metabolic disorders. Here, we provide a comprehensive overview of pathways and players involved in the development of brown and beige fat, as well as the role of thermogenic adipocytes in energy homeostasis and metabolism. Furthermore, we discuss the alterations in brown and beige adipose tissue function during obesity and explore the therapeutic potential of thermogenic activation to treat metabolic syndrome.
Collapse
Affiliation(s)
- Bruna B. Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Ankita Poojari
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| | - Atefeh Rabiee
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
28
|
Shinde AB, Song A, Wang QA. Brown Adipose Tissue Heterogeneity, Energy Metabolism, and Beyond. Front Endocrinol (Lausanne) 2021; 12:651763. [PMID: 33953697 PMCID: PMC8092391 DOI: 10.3389/fendo.2021.651763] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/18/2021] [Indexed: 01/19/2023] Open
Abstract
Brown adipocyte in brown adipose tissue (BAT) specializes in expending energy through non-shivering thermogenesis, a process that produces heat either by uncoupling protein 1 (UCP1) dependent uncoupling of mitochondrial respiration or by UCP1 independent mechanisms. Apart from this, there is ample evidence suggesting that BAT has an endocrine function. Studies in rodents point toward its vital roles in glucose and lipid homeostasis, making it an important therapeutic target for treating metabolic disorders related to morbidities such as obesity and type 2 diabetes. The rediscovery of thermogenically active BAT depots in humans by several independent research groups in the last decade has revitalized interest in BAT as an even more promising therapeutic intervention. Over the last few years, there has been overwhelming interest in understanding brown adipocyte's developmental lineages and how brown adipocyte uniquely utilizes energy beyond UCP1 mediated uncoupling respiration. These new discoveries would be leveraged for designing novel therapeutic interventions for metabolic disorders.
Collapse
Affiliation(s)
- Abhijit Babaji Shinde
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
| | - Anying Song
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
| | - Qiong A. Wang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
| |
Collapse
|
29
|
Hong H, Baatar D, Hwang SG. The difference of castration timing of Korean Hanwoo bulls does not significantly affect the carcass characteristics. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:426-439. [PMID: 33987616 PMCID: PMC8071752 DOI: 10.5187/jast.2021.e26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 11/20/2022]
Abstract
It is already well known that castration improves marbling quality but exact timing of castration is still highly debated in beef cattle production industry. After castration, blood hormonal changes occur in steer and objective of this study was to investigate the effects of growth hormone (GH) levels on adipocyte differentiation in stromal vascular cells (SVCs) and transdifferentiation into adipocytes in C2C12 myoblasts. Total GH concentrations were measured via enzyme-linked immunosorbent assay (ELISA) in 24 male calves and 4 female calves. Cell proliferation, cellular triglyceride (TG) accumulation, and the cell's lipolytic capability were measured in C2C12 myoblasts and SVCs. Myogenic, adipogenic, and brown adipocyte-specific gene expression was measured via real-time polymerase chain reaction (PCR) using SYBR green. Serum GH levels were the highest in late-castrated calves. Treatment with 5 ng/mL GH resulted in greater TG accumulation as well as increased CCAAT-enhancer-binding protein (C/EBP)α and peroxisome proliferator-activated receptor (PPAR)γ expression compared to that after treatment with 15 ng/mL GH. Treatment with 5 ng/mL GH also resulted in lower myogenin (myo)G and myoD expression compared to that after treatment with 15 ng/mL GH. The expression of bone morphogenetic protein (BMP) 7 after treatment with 5 ng/mL GH was higher than that after treatment with 15 ng/mL GH. But carcass characteristics data showed no significant difference between early and late castrated steers. Therefore, our results indicate that castration timing does not seem to be inevitable determinate of carcass qualities, particularly carcass weight and marbling score in Hanwoo beef cattle.
Collapse
Affiliation(s)
- Heeok Hong
- Department of Animal Science and
Technology, Konkuk University, Seoul 05029, Korea
| | - Delgerzul Baatar
- Laboratory of Genetics, Institute of
Biology, Peace Avenue, Bayanzurkh District, Ulaanbaatar 13330,
Mongolia
- School of Animal Life Convergence Science,
Hankyong National University, Anseong 17579, Korea
| | - Seong-Gu Hwang
- School of Animal Life Convergence Science,
Hankyong National University, Anseong 17579, Korea
| |
Collapse
|
30
|
Qian S, Tang Y, Tang QQ. Adipose tissue plasticity and the pleiotropic roles of BMP signaling. J Biol Chem 2021; 296:100678. [PMID: 33872596 PMCID: PMC8131923 DOI: 10.1016/j.jbc.2021.100678] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Adipose tissues, including white, beige, and brown adipose tissue, have evolved to be highly dynamic organs. Adipose tissues undergo profound changes during development and regeneration and readily undergo remodeling to meet the demands of an everchanging metabolic landscape. The dynamics are determined by the high plasticity of adipose tissues, which contain various cell types: adipocytes, immune cells, endothelial cells, nerves, and fibroblasts. There are numerous proteins that participate in regulating the plasticity of adipose tissues. Among these, bone morphogenetic proteins (BMPs) were initially found to regulate the differentiation of adipocytes, and they are being reported to have pleiotropic functions by emerging studies. Here, in the first half of the article, we summarize the plasticity of adipocytes and macrophages, which are two groups of cells targeted by BMP signaling in adipose tissues. We then review how BMPs regulate the differentiation, death, and lipid metabolism of adipocytes. In addition, the potential role of BMPs in regulating adipose tissue macrophages is considered. Finally, the expression of BMPs in adipose tissues and their metabolic relevance are discussed.
Collapse
Affiliation(s)
- Shuwen Qian
- The 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, China
| | - Yan Tang
- The 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, China
| | - Qi-Qun Tang
- The 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, China.
| |
Collapse
|
31
|
Thermogenic adipocytes: lineage, function and therapeutic potential. Biochem J 2020; 477:2071-2093. [PMID: 32539124 PMCID: PMC7293110 DOI: 10.1042/bcj20200298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
Metabolic inflexibility, defined as the inability to respond or adapt to metabolic demand, is now recognised as a driving factor behind many pathologies associated with obesity and the metabolic syndrome. Adipose tissue plays a pivotal role in the ability of an organism to sense, adapt to and counteract environmental changes. It provides a buffer in times of nutrient excess, a fuel reserve during starvation and the ability to resist cold-stress through non-shivering thermogenesis. Recent advances in single-cell RNA sequencing combined with lineage tracing, transcriptomic and proteomic analyses have identified novel adipocyte progenitors that give rise to specialised adipocytes with diverse functions, some of which have the potential to be exploited therapeutically. This review will highlight the common and distinct functions of well-known adipocyte populations with respect to their lineage and plasticity, as well as introducing the most recent members of the adipocyte family and their roles in whole organism energy homeostasis. Finally, this article will outline some of the more preliminary findings from large data sets generated by single-cell transcriptomics of mouse and human adipose tissue and their implications for the field, both for discovery and for therapy.
Collapse
|
32
|
Lizcano F, Arroyave F. Control of Adipose Cell Browning and Its Therapeutic Potential. Metabolites 2020; 10:metabo10110471. [PMID: 33227979 PMCID: PMC7699191 DOI: 10.3390/metabo10110471] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/20/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is the largest endocrine organ in humans and has an important influence on many physiological processes throughout life. An increasing number of studies have described the different phenotypic characteristics of fat cells in adults. Perhaps one of the most important properties of fat cells is their ability to adapt to different environmental and nutritional conditions. Hypothalamic neural circuits receive peripheral signals from temperature, physical activity or nutrients and stimulate the metabolism of white fat cells. During this process, changes in lipid inclusion occur, and the number of mitochondria increases, giving these cells functional properties similar to those of brown fat cells. Recently, beige fat cells have been studied for their potential role in the regulation of obesity and insulin resistance. In this context, it is important to understand the embryonic origin of beige adipocytes, the response of adipocyte to environmental changes or modifications within the body and their ability to transdifferentiate to elucidate the roles of these cells for their potential use in therapeutic strategies for obesity and metabolic diseases. In this review, we discuss the origins of the different fat cells and the possible therapeutic properties of beige fat cells.
Collapse
Affiliation(s)
- Fernando Lizcano
- Center of Biomedical Investigation, (CIBUS), Universidad de La Sabana, 250008 Chia, Colombia
- Correspondence:
| | - Felipe Arroyave
- Doctoral Program in Biociencias, Universidad de La Sabana, 250008 Chia, Colombia
| |
Collapse
|
33
|
Gharanei S, Shabir K, Brown JE, Weickert MO, Barber TM, Kyrou I, Randeva HS. Regulatory microRNAs in Brown, Brite and White Adipose Tissue. Cells 2020; 9:cells9112489. [PMID: 33207733 PMCID: PMC7696849 DOI: 10.3390/cells9112489] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/02/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) constitute a class of short noncoding RNAs which regulate gene expression by targeting messenger RNA, inducing translational repression and messenger RNA degradation. This regulation of gene expression by miRNAs in adipose tissue (AT) can impact on the regulation of metabolism and energy homeostasis, particularly considering the different types of adipocytes which exist in mammals, i.e., white adipocytes (white AT; WAT), brown adipocytes (brown AT; BAT), and inducible brown adipocytes in WAT (beige or brite or brown-in-white adipocytes). Indeed, an increasing number of miRNAs has been identified to regulate key signaling pathways of adipogenesis in BAT, brite AT, and WAT by acting on transcription factors that promote or inhibit adipocyte differentiation. For example, MiR-328, MiR-378, MiR-30b/c, MiR-455, MiR-32, and MiR-193b-365 activate brown adipogenesis, whereas MiR-34a, MiR-133, MiR-155, and MiR-27b are brown adipogenesis inhibitors. Given that WAT mainly stores energy as lipids, whilst BAT mainly dissipates energy as heat, clarifying the effects of miRNAs in different types of AT has recently attracted significant research interest, aiming to also develop novel miRNA-based therapies against obesity, diabetes, and other obesity-related diseases. Therefore, this review presents an up-to-date comprehensive overview of the role of key regulatory miRNAs in BAT, brite AT, and WAT.
Collapse
Affiliation(s)
- Seley Gharanei
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Kiran Shabir
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
| | - James E. Brown
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre of Applied Biological & Exercise Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
- Correspondence:
| |
Collapse
|
34
|
Zhao Q, Zhang Z, Rong W, Jin W, Yan L, Jin W, Xu Y, Cui X, Tang QQ, Pan D. KMT5c modulates adipocyte thermogenesis by regulating Trp53 expression. Proc Natl Acad Sci U S A 2020; 117:22413-22422. [PMID: 32839323 PMCID: PMC7486735 DOI: 10.1073/pnas.1922548117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Brown and beige adipocytes harbor the thermogenic capacity to adapt to environmental thermal or nutritional changes. Histone methylation is an essential epigenetic modification involved in the modulation of nonshivering thermogenesis in adipocytes. Here, we describe a molecular network leading by KMT5c, a H4K20 methyltransferase, that regulates adipocyte thermogenesis and systemic energy expenditure. The expression of Kmt5c is dramatically induced by a β3-adrenergic signaling cascade in both brown and beige fat cells. Depleting Kmt5c in adipocytes in vivo leads to a decreased expression of thermogenic genes in both brown and subcutaneous (s.c.) fat tissues. These mice are prone to high-fat-diet-induced obesity and develop glucose intolerance. Enhanced transformation related protein 53 (Trp53) expression in Kmt5c knockout (KO) mice, that is due to the decreased repressive mark H4K20me3 on its proximal promoter, is responsible for the metabolic phenotypes. Together, these findings reveal the physiological role for KMT5c-mediated H4K20 methylation in the maintenance and activation of the thermogenic program in adipocytes.
Collapse
Affiliation(s)
- Qingwen Zhao
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of China
| | - Zhe 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, Fudan University, 200032 Shanghai, People's Republic of China
| | - Weiqiong Rong
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of China
| | - Weiwei Jin
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of China
| | - Linyu Yan
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of China
| | - Wenfang Jin
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of China
| | - Yingjiang Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of China
| | - Xuan Cui
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of 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, Fudan University, 200032 Shanghai, People's Republic of China
| | - Dongning Pan
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, 200032 Shanghai, People's Republic of China
| |
Collapse
|
35
|
Zhang X, Wang X, Yin H, Zhang L, Feng A, Zhang QX, Lin Y, Bao B, Hernandez LL, Shi GP, Liu J. Functional Inactivation of Mast Cells Enhances Subcutaneous Adipose Tissue Browning in Mice. Cell Rep 2020; 28:792-803.e4. [PMID: 31315055 DOI: 10.1016/j.celrep.2019.06.044] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 04/08/2019] [Accepted: 06/12/2019] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue browning and systemic energy expenditure provide a defense mechanism against obesity and associated metabolic diseases. In high-cholesterol Western diet-fed mice, mast cell (MC) inactivation ameliorates obesity and insulin resistance and improves the metabolic rate, but a direct role of adipose tissue MCs in thermogenesis and browning remains unproven. Here, we report that adrenoceptor agonist norepinephrine-stimulated metabolic rate and subcutaneous adipose tissue (SAT) browning are enhanced in MC-deficient Kitw-sh/w-sh mice and MC-stabilized wild-type mice on a chow diet. MC reconstitution to SAT in Kitw-sh/w-sh mice blocks these changes. Mechanistic studies demonstrate that MC inactivation elevates SAT platelet-derived growth factor receptor A (PDGFRα+) adipocyte precursor proliferation and accelerates beige adipocyte differentiation. Using the tryptophan hydroxylase 1 (TPH1) inhibitor and TPH1-deficient MCs, we show that MC-derived serotonin inhibits SAT browning and systemic energy expenditure. Functional inactivation of MCs or inhibition of MC serotonin synthesis in SAT promotes adipocyte browning and systemic energy metabolism in mice.
Collapse
Affiliation(s)
- Xian Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Xin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Hao Yin
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Lei Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Airong Feng
- Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Qiu-Xia Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yan Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Bin Bao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
| | - Laura L Hernandez
- Department of Dairy Science, University of Wisconsin, Madison, WI 53706, USA
| | - Guo-Ping Shi
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230009, China.
| |
Collapse
|
36
|
Ambele MA, Dhanraj P, Giles R, Pepper MS. Adipogenesis: A Complex Interplay of Multiple Molecular Determinants and Pathways. Int J Mol Sci 2020; 21:E4283. [PMID: 32560163 PMCID: PMC7349855 DOI: 10.3390/ijms21124283] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/07/2020] [Indexed: 11/24/2022] Open
Abstract
The formation of adipocytes during embryogenesis has been largely understudied. However, preadipocytes appear to originate from multipotent mesenchymal stromal/stem cells which migrate from the mesoderm to their anatomical localization. Most studies on adipocyte formation (adipogenesis) have used preadipocytes derived from adult stem/stromal cells. Adipogenesis consists of two phases, namely commitment and terminal differentiation. This review discusses the role of signalling pathways, epigenetic modifiers, and transcription factors in preadipocyte commitment and differentiation into mature adipocytes, as well as limitations in our understanding of these processes. To date, a limited number of transcription factors, genes and signalling pathways have been described to regulate preadipocyte commitment. One reason could be that most studies on adipogenesis have used preadipocytes already committed to the adipogenic lineage, which are therefore not suitable for studying preadipocyte commitment. Conversely, over a dozen molecular players including transcription factors, genes, signalling pathways, epigenetic regulators, and microRNAs have been described to be involved in the differentiation of preadipocytes to adipocytes; however, only peroxisome proliferator-activated receptor gamma has proven to be clinically relevant. A detailed understanding of how the molecular players underpinning adipogenesis relate to adipose tissue function could provide new therapeutic approaches for addressing obesity without compromising adipose tissue function.
Collapse
Affiliation(s)
- Melvin A. Ambele
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Priyanka Dhanraj
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
| | - Rachel Giles
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
| | - Michael S. Pepper
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
| |
Collapse
|
37
|
Single-cell transcriptional networks in differentiating preadipocytes suggest drivers associated with tissue heterogeneity. Nat Commun 2020; 11:2117. [PMID: 32355218 PMCID: PMC7192917 DOI: 10.1038/s41467-020-16019-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
White adipose tissue plays an important role in physiological homeostasis and metabolic disease. Different fat depots have distinct metabolic and inflammatory profiles and are differentially associated with disease risk. It is unclear whether these differences are intrinsic to the pre-differentiated stage. Using single-cell RNA sequencing, a unique network methodology and a data integration technique, we predict metabolic phenotypes in differentiating cells. Single-cell RNA-seq profiles of human preadipocytes during adipogenesis in vitro identifies at least two distinct classes of subcutaneous white adipocytes. These differences in gene expression are separate from the process of browning and beiging. Using a systems biology approach, we identify a new network of zinc-finger proteins that are expressed in one class of preadipocytes and is potentially involved in regulating adipogenesis. Our findings gain a deeper understanding of both the heterogeneity of white adipocytes and their link to normal metabolism and disease. The origin of the heterogeneity of metabolic and inflammatory profiles exhibited by white adipocytes is little understood. Here, using scRNA-seq and computational methods, the authors show that differentiating preadipocytes exhibit gene expression differences and suggest underlying regulators.
Collapse
|
38
|
Xu Z, You W, Liu J, Wang Y, Shan T. Elucidating the Regulatory Role of Melatonin in Brown, White, and Beige Adipocytes. Adv Nutr 2020; 11:447-460. [PMID: 31355852 PMCID: PMC7442421 DOI: 10.1093/advances/nmz070] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/28/2019] [Accepted: 06/13/2019] [Indexed: 12/15/2022] Open
Abstract
The high prevalence of obesity and its associated metabolic diseases has heightened the importance of understanding control of adipose tissue development and energy metabolism. In mammals, 3 types of adipocytes with different characteristics and origins have been identified: white, brown, and beige. Beige and brown adipocytes contain numerous mitochondria and have the capability to burn energy and counteract obesity, while white adipocytes store energy and are closely associated with metabolic disorders and obesity. Thus, regulation of the development and function of different adipocytes is important for controlling energy balance and combating obesity and related metabolic disorders. Melatonin is a neurohormone, which plays multiple roles in regulating inflammation, blood pressure, insulin actions, and energy metabolism. This article summarizes and discusses the role of melatonin in white, beige, and brown adipocytes, especially in affecting adipogenesis, inducing beige formation or white adipose tissue browning, enhancing brown adipose tissue mass and activities, improving anti-inflammatory and antioxidative effects, regulating adipokine secretion, and preventing body weight gain. Based on the current findings, melatonin is a potential therapeutic agent to control energy metabolism, adipogenesis, fat deposition, adiposity, and related metabolic diseases.
Collapse
Affiliation(s)
- Ziye Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China; and Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China,Address correspondence to TS (E-mail: )
| |
Collapse
|
39
|
Seoane-Collazo P, Martínez-Sánchez N, Milbank E, Contreras C. Incendiary Leptin. Nutrients 2020; 12:nu12020472. [PMID: 32069871 PMCID: PMC7071158 DOI: 10.3390/nu12020472] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/06/2020] [Accepted: 02/08/2020] [Indexed: 02/08/2023] Open
Abstract
Leptin is a hormone released by adipose tissue that plays a key role in the control of energy homeostasis through its binding to leptin receptors (LepR), mainly expressed in the hypothalamus. Most scientific evidence points to leptin’s satiating effect being due to its dual capacity to promote the expression of anorexigenic neuropeptides and to reduce orexigenic expression in the hypothalamus. However, it has also been demonstrated that leptin can stimulate (i) thermogenesis in brown adipose tissue (BAT) and (ii) the browning of white adipose tissue (WAT). Since the demonstration of the importance of BAT in humans 10 years ago, its study has aroused great interest, mainly in the improvement of obesity-associated metabolic disorders through the induction of thermogenesis. Consequently, several strategies targeting BAT activation (mainly in rodent models) have demonstrated great potential to improve hyperlipidemias, hepatic steatosis, insulin resistance and weight gain, leading to an overall healthier metabolic profile. Here, we review the potential therapeutic ability of leptin to correct obesity and other metabolic disorders, not only through its satiating effect, but by also utilizing its thermogenic properties.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Noelia Martínez-Sánchez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Edward Milbank
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Cristina Contreras
- Department of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| |
Collapse
|
40
|
Abstract
Understanding the mammalian energy balance can pave the way for future therapeutics that enhance energy expenditure as an anti-obesity and anti-diabetic strategy. Several studies showed that brown adipose tissue activity increases daily energy expenditure. However, the size and activity of brown adipose tissue is reduced in individuals with obesity and type two diabetes. Humans have an abundance of functionally similar beige adipocytes that have the potential to contribute to increased energy expenditure. This makes beige adipocytes a promising target for metabolic disease therapies. While brown adipocytes tend to be stable, beige adipocytes have a high level of plasticity that allows for the rapid and dynamic induction of thermogenesis by external stimuli such as low environmental temperatures. This means that after browning stimuli have been withdrawn beige adipocytes quickly transition back to their white adipose state. The detailed molecular mechanisms regulating beige adipocytes development, function, and reversibility are not fully understood. The goal of this review is to give a comprehensive overview of beige fat development and origins, along with the transcriptional and epigenetic programs that lead to beige fat formation, and subsequent thermogenesis in humans. An improved understanding of the molecular pathways of beige adipocyte plasticity will enable us to selectively manipulate beige cells to induce and maintain their thermogenic output thus improving the whole-body energy homeostasis.
Collapse
|
41
|
Lizcano F. The Beige Adipocyte as a Therapy for Metabolic Diseases. Int J Mol Sci 2019; 20:ijms20205058. [PMID: 31614705 PMCID: PMC6834159 DOI: 10.3390/ijms20205058] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 09/30/2019] [Accepted: 10/05/2019] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue is traditionally categorized into white and brown relating to their function and morphology. The classical white adipose tissue builds up energy in the form of triglycerides and is useful for preventing fatigue during periods of low caloric intake and the brown adipose tissue more energetically active, with a greater number of mitochondria and energy production in the form of heat. Since adult humans possess significant amounts of active brown fat depots and its mass inversely correlates with adiposity, brown fat might play an important role in human obesity and energy homeostasis. New evidence suggests two types of thermogenic adipocytes with distinct developmental and anatomical features: classical brown adipocytes and beige adipocytes. Beige adipocyte has recently attracted special interest because of its ability to dissipate energy and the possible ability to differentiate themselves from white adipocytes. The presence of brown and beige adipocyte in human adults has acquired attention as a possible therapeutic intervention for metabolic diseases. Importantly, adult human brown appears to be mainly composed of beige-like adipocytes, making this cell type an attractive therapeutic target for obesity and obesity-related diseases, such as atherosclerosis, arterial hypertension and diabetes mellitus type 2. Because many epigenetics changes can affect beige adipocyte differentiation from adipose progenitor cells, the knowledge of the circumstances that affect the development of beige adipocyte cells may be important to new pathways in the treatment of metabolic diseases. New molecules have emerged as possible therapeutic targets, which through the impulse to develop beige adipocytes can be useful for clinical studies. In this review will discuss some recent observations arising from the unique physiological capacity of these cells and their possible role as ways to treat obesity and diabetes mellitus type 2.
Collapse
Affiliation(s)
- Fernando Lizcano
- Center of Biomedical Investigation, (CIBUS), Universidad de La Sabana, 250008 Chia, Colombia.
| |
Collapse
|
42
|
Brown AC. Brown adipocytes from induced pluripotent stem cells-how far have we come? Ann N Y Acad Sci 2019; 1463:9-22. [PMID: 31573081 DOI: 10.1111/nyas.14257] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
A global increase in the number of individuals who are either overweight or obese is leading to a higher incidence of type 2 diabetes (T2D). Behavioral interventions for the treatment of obesity have yet to deliver desired outcomes, thus introducing a pressing need for molecular- and cellular-based therapies. Excess energy from food is stored in the form of triglycerides in white adipose tissue, which expands during weight gain and can lead to insulin resistance and T2D. By contrast, brown adipose tissue (BAT) releases energy from metabolic substrates in the form of heat and secretes factors that can reverse metabolic disease by acting systemically. Therefore, the ability to increase BAT activity is a promising approach to improve energy balance and metabolic homeostasis. Methods are now being developed to generate brown adipocytes from human induced pluripotent stem cells (hiPSCs), which would (1) provide an unlimited source of cellular material to study human brown adipogenesis, and (2) could be used to develop drug- and cell-based therapies for the treatment of metabolic complications associated with obesity. This article reviews basic BAT biology and details the current progress toward developing brown adipocytes from hiPSCs.
Collapse
Affiliation(s)
- Aaron C Brown
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine.,School of Biomedical Sciences and Engineering, The University of Maine, Orono, Maine.,Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
43
|
Jung SM, Sanchez-Gurmaches J, Guertin DA. Brown Adipose Tissue Development and Metabolism. Handb Exp Pharmacol 2019; 251:3-36. [PMID: 30203328 DOI: 10.1007/164_2018_168] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Brown adipose tissue is well known to be a thermoregulatory organ particularly important in small rodents and human infants, but it was only recently that its existence and significance to metabolic fitness in adult humans have been widely realized. The ability of active brown fat to expend high amounts of energy has raised interest in stimulating thermogenesis therapeutically to treat metabolic diseases related to obesity and type 2 diabetes. In parallel, there has been a surge of research aimed at understanding the biology of rodent and human brown fat development, its remarkable metabolic properties, and the phenomenon of white fat browning, in which white adipocytes can be converted into brown like adipocytes with similar thermogenic properties. Here, we review the current understanding of the developmental and metabolic pathways involved in forming thermogenic adipocytes, and highlight some of the many unknown functions of brown fat that make its study a rich and exciting area for future research.
Collapse
Affiliation(s)
- Su Myung Jung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA. .,Molecular, Cell and Cancer Biology Program, University of Massachusetts Medical School, Worcester, MA, USA. .,Lei Weibo Institute for Rare Diseases, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
44
|
Zhu Q, Glazier BJ, Hinkel BC, Cao J, Liu L, Liang C, Shi H. Neuroendocrine Regulation of Energy Metabolism Involving Different Types of Adipose Tissues. Int J Mol Sci 2019; 20:E2707. [PMID: 31159462 PMCID: PMC6600468 DOI: 10.3390/ijms20112707] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 12/17/2022] Open
Abstract
Despite tremendous research efforts to identify regulatory factors that control energy metabolism, the prevalence of obesity has been continuously rising, with nearly 40% of US adults being obese. Interactions between secretory factors from adipose tissues and the nervous system innervating adipose tissues play key roles in maintaining energy metabolism and promoting survival in response to metabolic challenges. It is currently accepted that there are three types of adipose tissues, white (WAT), brown (BAT), and beige (BeAT), all of which play essential roles in maintaining energy homeostasis. WAT mainly stores energy under positive energy balance, while it releases fuels under negative energy balance. Thermogenic BAT and BeAT dissipate energy as heat under cold exposure to maintain body temperature. Adipose tissues require neural and endocrine communication with the brain. A number of WAT adipokines and BAT batokines interact with the neural circuits extending from the brain to cooperatively regulate whole-body lipid metabolism and energy homeostasis. We review neuroanatomical, histological, genetic, and pharmacological studies in neuroendocrine regulation of adipose function, including lipid storage and mobilization of WAT, non-shivering thermogenesis of BAT, and browning of BeAT. Recent whole-tissue imaging and transcriptome analysis of differential gene expression in WAT and BAT yield promising findings to better understand the interaction between secretory factors and neural circuits, which represents a novel opportunity to tackle obesity.
Collapse
Affiliation(s)
- Qi Zhu
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Bradley J Glazier
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Benjamin C Hinkel
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Jingyi Cao
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Lin Liu
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Chun Liang
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Haifei Shi
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
45
|
Grigoraş A, Amalinei C, Balan RA, Giuşcă SE, Avădănei ER, Lozneanu L, Căruntu ID. Adipocytes spectrum - From homeostasia to obesity and its associated pathology. Ann Anat 2018; 219:102-120. [PMID: 30049662 DOI: 10.1016/j.aanat.2018.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 06/17/2018] [Indexed: 02/07/2023]
Abstract
Firstly identified by anatomists, the fat tissue is nowadays an area of intense research due to increased global prevalence of obesity and its associated diseases. Histologically, there are four types of fat tissue cells which are currently recognized (white, brown, beige, and perivascular adipocytes). Therefore, in this study we are reviewing the most recent data regarding the origin, structure, and molecular mechanisms involved in the development of adipocytes. White adipocytes can store triglycerides as a consequence of lipogenesis, under the regulation of growth hormone or leptin and adiponectin, and release fatty acids resulted from lipolysis, under the regulation of the sympathetic nervous system, glucocorticoids, TNF-α, insulin, and natriuretic peptides. Brown adipocytes possess a mitochondrial transmembrane protein thermogenin or UCP1 which allows heat generation. Recently, thermogenic, UCP positive adipocytes have been identified in the subcutaneous white adipose tissue and have been named beige adipocytes. The nature of these cells is still controversial, as current theories are suggesting their origin either by transdifferentiation of white adipocytes, or by differentiation from an own precursor cell. Perivascular adipocytes surround most of the arteries, exhibiting a supportive role and being involved in the maintenance of intravascular temperature. Thoracic perivascular adipocytes resemble brown adipocytes, while abdominal ones are more similar to white adipocytes and, consequently, are involved in obesity-induced inflammatory reactions. The factors involved in the regulation of adipose stem cells differentiation may represent potential pathways to inhibit or to divert adipogenesis. Several molecules, such as pro-adipogenic factors (FGF21, BMP7, BMP8b, and Cox-2), cell surface proteins or receptors (Asc-1, PAT2, P2RX5), and hypothalamic receptors (MC4R) have been identified as the most promising targets for the development of future therapies. Further investigations are necessary to complete the knowledge about adipose tissue and the development of a new generation of therapeutic tools based on molecular targets.
Collapse
Affiliation(s)
- Adriana Grigoraş
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania; Department of Histopathology, Institute of Legal Medicine, Iasi, Romania.
| | - Cornelia Amalinei
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania; Department of Histopathology, Institute of Legal Medicine, Iasi, Romania.
| | - Raluca Anca Balan
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Simona Eliza Giuşcă
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Elena Roxana Avădănei
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Ludmila Lozneanu
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Irina-Draga Căruntu
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| |
Collapse
|
46
|
Schoettl T, Fischer IP, Ussar S. Heterogeneity of adipose tissue in development and metabolic function. ACTA ACUST UNITED AC 2018. [PMID: 29514879 DOI: 10.1242/jeb.162958] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adipose tissue is a central metabolic organ. Unlike other organs, adipose tissue is compartmentalized into individual depots and distributed throughout the body. These different adipose depots show major functional differences and risk associations for developing metabolic syndrome. Recent advances in lineage tracing demonstrate that individual adipose depots are composed of adipocytes that are derived from distinct precursor populations, giving rise to different populations of energy-storing white adipocytes. Moreover, distinct lineages of energy-dissipating brown and beige adipocytes exist in discrete depots or within white adipose tissue depots. In this Review, we discuss developmental and functional heterogeneity, as well as sexual dimorphism, between and within individual adipose tissue depots. We highlight current data relating to the differences between subcutaneous and visceral white adipose tissue in the development of metabolic dysfunction, with special emphasis on adipose tissue expansion and remodeling of the extracellular matrix. Moreover, we provide a detailed overview of adipose tissue development as well as the consensus and controversies relating to adult adipocyte precursor populations.
Collapse
Affiliation(s)
- Theresa Schoettl
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ingrid P Fischer
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.,Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Siegfried Ussar
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany .,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|
47
|
Lynes MD, Tseng YH. Deciphering adipose tissue heterogeneity. Ann N Y Acad Sci 2018; 1411:5-20. [PMID: 28763833 PMCID: PMC5788721 DOI: 10.1111/nyas.13398] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/02/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023]
Abstract
Obesity is an excess accumulation of adipose tissue mass, and, together with its sequelae, in particular type II diabetes and metabolic syndrome, obesity presents a major health crisis. Although obesity is simply caused by increased adipose mass, the heterogeneity of adipose tissue in humans means that the response to increased energy balance is highly complex. Individual subjects with similar phenotypes may respond very differently to the same treatments; therefore, obesity may benefit from a personalized precision medicine approach. The variability in the development of obesity is indeed driven by differences in sex, genetics, and environment, but also by the various types of adipose tissue as well as the different cell types that compose it. By describing the distinct cell populations that reside in different fat depots, we can interpret the complex effect of these various players in the maintenance of whole-body energy homeostasis. To further understand adipose tissue, adipogenic differentiation and the transcriptional program of lipid accumulation must be investigated. As the cell- and depot-specific functions are described, they can be placed in the context of energy excess to understand how the heterogeneity of adipose tissue shapes individual metabolic status and condition.
Collapse
Affiliation(s)
- Matthew D Lynes
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
48
|
Barrott JJ, Illum BE, Jin H, Hedberg ML, Wang Y, Grossmann A, Haldar M, Capecchi MR, Jones KB. Paracrine osteoprotegerin and β-catenin stabilization support synovial sarcomagenesis in periosteal cells. J Clin Invest 2017; 128:207-218. [PMID: 29202462 DOI: 10.1172/jci94955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/10/2017] [Indexed: 01/30/2023] Open
Abstract
Synovial sarcoma (SS) is an aggressive soft-tissue sarcoma that is often discovered during adolescence and young adulthood. Despite the name, synovial sarcoma does not typically arise from a synoviocyte but instead arises in close proximity to bones. Previous work demonstrated that mice expressing the characteristic SS18-SSX fusion oncogene in myogenic factor 5-expressing (Myf5-expressing) cells develop fully penetrant sarcomagenesis, suggesting skeletal muscle progenitor cell origin. However, Myf5 is not restricted to committed myoblasts in embryos but is also expressed in multipotent mesenchymal progenitors. Here, we demonstrated that human SS and mouse tumors arising from SS18-SSX expression in the embryonic, but not postnatal, Myf5 lineage share an anatomic location that is frequently adjacent to bone. Additionally, we showed that SS can originate from periosteal cells expressing SS18-SSX alone and from preosteoblasts expressing the fusion oncogene accompanied by the added stabilization of β-catenin, which is a common secondary change in SS. Expression and secretion of the osteoclastogenesis inhibitory factor osteoprotegerin enabled early growth of SS18-SSX2-transformed cells, indicating a paracrine link between the bone and synovial sarcomagenesis. These findings explain the skeletal contact frequently observed in human SS and may provide alternate means of enabling SS18-SSX-driven oncogenesis in cells as differentiated as preosteoblasts.
Collapse
Affiliation(s)
| | - Benjamin E Illum
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Huifeng Jin
- Departments of Orthopaedics and Oncological Sciences, and
| | - Matthew L Hedberg
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
| | - Yanliang Wang
- Departments of Orthopaedics and Oncological Sciences, and
| | - Allie Grossmann
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Malay Haldar
- Department of Pathology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mario R Capecchi
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Kevin B Jones
- Departments of Orthopaedics and Oncological Sciences, and
| |
Collapse
|
49
|
Okamatsu-Ogura Y, Nio-Kobayashi J, Nagaya K, Tsubota A, Kimura K. Brown adipocytes postnatally arise through both differentiation from progenitors and conversion from white adipocytes in Syrian hamster. J Appl Physiol (1985) 2017; 124:99-108. [PMID: 28982944 DOI: 10.1152/japplphysiol.00622.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To investigate the postnatal development of brown adipose tissue (BAT) in Syrian hamsters, we histologically examined interscapular fat tissue from 5-16-day-old pups, focusing on how brown adipocytes arise. Interscapular fat of 5-day-old hamsters mainly consisted of white adipocytes containing large unilocular lipid droplets, as observed in typical white adipose tissue (WAT). On day 7, clusters of small, proliferative nonadipocytes with a strong immunoreactivity for Ki67 appeared near the edge of the interscapular fat tissue. The area of the Ki67-positive regions expanded to ~50% of the total tissue area by day 10. The interscapular fat showed the typical BAT feature by day 16. A brown adipocyte-specific marker, uncoupling protein-1, was clearly detected on day 10 and thereafter, while not detected on day 7. During conversion of interscapular fat from WAT to BAT, unilocular adipocytes completely and rapidly disappeared without obvious apoptosis. Dual immunofluorescence staining for Ki67 and monocarboxylate transporter 1 (MCT1), another selective marker for brown adipocytes, revealed that most of the proliferating cells were of the brown adipocyte lineage. Electron microscopic examination showed that some of the white adipocytes contained small lipid droplets in addition to the large droplet and expressed MCT1 as do progenitor and mature brown adipocytes, implying a direct conversion from white to brown adipocytes. These results suggest that BAT of Syrian hamsters develops postnatally through two different pathways: the proliferation and differentiation of brown adipocyte progenitors and the conversion of unilocular adipocytes to multilocular brown adipocytes. NEW & NOTEWORTHY Brown and white adipose tissues (BAT and WAT, respectively) are quite different in morphological features and function; however, the boundary between these tissues is obscure. In this study, we histologically evaluated the process of BAT development in Syrian hamsters, which shows postnatal conversion of WAT to BAT. Our results suggest that brown adipocytes arise through two different pathways: the proliferation and differentiation of brown adipocyte progenitors and the conversion from white adipocytes.
Collapse
Affiliation(s)
- Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| | - Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University , Sapporo , Japan
| | - Kazuki Nagaya
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| | - Ayumi Tsubota
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| | - Kazuhiro Kimura
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| |
Collapse
|
50
|
Jankovic A, Otasevic V, Stancic A, Buzadzic B, Korac A, Korac B. Physiological regulation and metabolic role of browning in white adipose tissue. Horm Mol Biol Clin Investig 2017; 31:hmbci-2017-0034. [PMID: 28862984 DOI: 10.1515/hmbci-2017-0034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 07/24/2017] [Indexed: 04/25/2024]
Abstract
Great progress has been made in our understanding of the browning process in white adipose tissue (WAT) in rodents. The recognition that i) adult humans have physiologically inducible brown adipose tissue (BAT) that may facilitate resistance to obesity and ii) that adult human BAT molecularly and functionally resembles beige adipose tissue in rodents, reignited optimism that obesity and obesity-related diabetes type 2 can be battled by controlling the browning of WAT. In this review the main cellular mechanisms and molecular mediators of browning of WAT in different physiological states are summarized. The relevance of browning of WAT in metabolic health is considered primarily through a modulation of biological role of fat tissue in overall metabolic homeostasis.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Vesna Otasevic
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Ana Stancic
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Biljana Buzadzic
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Aleksandra Korac
- Faculty of Biology, Center for Electron Microscopy, University of Belgrade, Belgrade, Serbia
| | - Bato Korac
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia, Phone: (381-11)-2078-307, Fax: (381-11)-2761-433
| |
Collapse
|