101
|
Interscapular and Perivascular Brown Adipose Tissue Respond Differently to a Short-Term High-Fat Diet. Nutrients 2019; 11:nu11051065. [PMID: 31086124 PMCID: PMC6566556 DOI: 10.3390/nu11051065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT) function may depend on its anatomical location and developmental origin. Interscapular BAT (iBAT) regulates acute macronutrient metabolism, whilst perivascular BAT (PVAT) regulates vascular function. Although phenotypically similar, whether these depots respond differently to acute nutrient excess is unclear. Given their distinct anatomical locations and developmental origins and we hypothesised that iBAT and PVAT would respond differently to brief period of nutrient excess. Sprague-Dawley rats aged 12 weeks (n=12) were fed either a standard (10% fat, n=6) or high fat diet (HFD: 45% fat, n=6) for 72h and housed at thermoneutrality. Following an assessment of whole body physiology, fat was collected from both depots for analysis of gene expression and the proteome. HFD consumption for 72h induced rapid weight gain (c. 2.6%) and reduced serum non-esterified fatty acids (NEFA) with no change in either total adipose or depot mass. In iBAT, an upregulation of genes involved in insulin signalling and lipid metabolism was accompanied by enrichment of lipid-related processes and functions, plus glucagon and peroxisome proliferator-activated receptor (PPAR) signalling pathways. In PVAT, HFD induced a pronounced down-regulation of multiple metabolic pathways which was accompanied with increased abundance of proteins involved in apoptosis (e.g., Hdgf and Ywaq) and toll-like receptor signalling (Ube2n). There was also an enrichment of DNA-related processes and functions (e.g., nucleosome assembly and histone exchange) and RNA degradation and cell adhesion pathways. In conclusion, we show that iBAT and PVAT elicit divergent responses to short-term nutrient excess highlighting early adaptations in these depots before changes in fat mass.
Collapse
|
102
|
Muller S, Ader I, Creff J, Leménager H, Achard P, Casteilla L, Sensebé L, Carrière A, Deschaseaux F. Human adipose stromal-vascular fraction self-organizes to form vascularized adipose tissue in 3D cultures. Sci Rep 2019; 9:7250. [PMID: 31076601 PMCID: PMC6510792 DOI: 10.1038/s41598-019-43624-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/18/2019] [Indexed: 12/17/2022] Open
Abstract
Native human subcutaneous adipose tissue (AT) is well organized into unilocular adipocytes interspersed within dense vascularization. This structure is completely lost under standard culture conditions and may impair the comparison with native tissue. Here, we developed a 3-D model of human white AT reminiscent of the cellular architecture found in vivo. Starting with adipose progenitors derived from the stromal-vascular fraction of human subcutaneous white AT, we generated spheroids in which endogenous endothelial cells self-assembled to form highly organized endothelial networks among stromal cells. Using an optimized adipogenic differentiation medium to preserve endothelial cells, we obtained densely vascularized spheroids containing mature adipocytes with unilocular lipid vacuoles. In vivo study showed that when differentiated spheroids were transplanted in immune-deficient mice, endothelial cells within the spheroids connected to the recipient circulatory system, forming chimeric vessels. In addition, adipocytes of human origin were still observed in transplanted mice. We therefore have developed an in vitro model of vascularized human AT-like organoids that constitute an excellent tool and model for any study of human AT.
Collapse
Affiliation(s)
- Sandra Muller
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Isabelle Ader
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Justine Creff
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France.,LBCMCP, Centre de Biologie Intégrative (CBI) CNRS, University of Toulouse, Toulouse, France.,LAAS-CNRS University of Toulouse CNRS, Toulouse, France
| | - Hélène Leménager
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Pauline Achard
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Louis Casteilla
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Luc Sensebé
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Audrey Carrière
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Frédéric Deschaseaux
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France.
| |
Collapse
|
103
|
Sebo ZL, Rodeheffer MS. Assembling the adipose organ: adipocyte lineage segregation and adipogenesis in vivo. Development 2019; 146:dev172098. [PMID: 30948523 PMCID: PMC6467474 DOI: 10.1242/dev.172098] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adipose tissue is composed of anatomically distinct depots that mediate several important aspects of energy homeostasis. The past two decades have witnessed increased research effort to elucidate the ontogenetic basis of adipose form and function. In this Review, we discuss advances in our understanding of adipose tissue development with particular emphasis on the embryonic patterning of depot-specific adipocyte lineages and adipocyte differentiation in vivo Micro-environmental cues and other factors that influence cell identity and cell behavior at various junctures in the adipocyte lineage hierarchy are also considered.
Collapse
Affiliation(s)
- Zachary L Sebo
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520-8103, USA
| | - Matthew S Rodeheffer
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520-8103, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520-8016, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520-8073, USA
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
104
|
Ramirez AK, Dankel S, Cai W, Sakaguchi M, Kasif S, Kahn CR. Membrane metallo-endopeptidase (Neprilysin) regulates inflammatory response and insulin signaling in white preadipocytes. Mol Metab 2019; 22:21-36. [PMID: 30795914 PMCID: PMC6437599 DOI: 10.1016/j.molmet.2019.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/04/2019] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Accumulation of visceral white adipose tissue (WAT) associates with insulin resistance, adipose tissue inflammation, and metabolic syndrome, whereas accumulation of subcutaneous WAT may be protective. We aimed to identify molecular mechanisms that might provide mechanistic insights underlying the phenotypic differences in these tissues. Membrane Metallo-Endopeptidase (MME/Neprislyin) is an extracellular, membrane-bound protease enriched in subcutaneous WAT that can target degradation of a variety of peptides, including insulin, IL6, and β-amyloids. We hypothesized that MME contributes to adipose depot-specific metabolic properties. METHODS We performed RNA sequencing on human subcutaneous and visceral preadipocytes and array gene expression profiling in murine subcutaneous and visceral preadipocytes. We conducted several insulin signaling and inflammatory response experiments on different cellular states of MME expression. RESULTS MME in white preadipocytes is expressed at a higher level in subcutaneous compared to visceral WAT and favors insulin signaling and a low inflammatory response. Thus, knockdown of MME in subcutaneous preadipocytes increased the inflammatory response to substance P and amyloid β aggregates. This associated with increased basal insulin signaling and decreased insulin-stimulated signaling. Moreover, MME differentially regulates the internalization and turnover of the α/β subunits of the insulin receptor. CONCLUSION MME is a novel regulator of the insulin receptor in adipose tissue. Given the clinical significance of both chronic inflammation and insulin sensitivity in metabolic disease, these results show a potentially new target to increase insulin sensitivity and decrease inflammatory susceptibility.
Collapse
Affiliation(s)
- Alfred K Ramirez
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Simon Dankel
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Hormone Laboratory, Haukeland University Hospital, 5020 Bergen, Norway
| | - Weikang Cai
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Masaji Sakaguchi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Simon Kasif
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Graduate Program in Bioinformatics, Boston University, Boston, MA 02215, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
105
|
Xie P, Zhao C, Huang W, Yong T, Chung ACK, He K, Chen X, Cai Z. Prenatal exposure to ambient fine particulate matter induces dysregulations of lipid metabolism in adipose tissue in male offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 657:1389-1397. [PMID: 30677905 DOI: 10.1016/j.scitotenv.2018.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/29/2018] [Accepted: 12/02/2018] [Indexed: 06/09/2023]
Abstract
Prenatal exposure to ambient fine particles (diameter < 0.25 μm, PM2.5) has been found to be associated with abnormal growth and development in offspring. However, the effects of PM2.5 on the lipid metabolism of adipose tissue in offspring are unclear. In the present study, we established a mouse model of prenatal exposure to PM2.5 by intratracheal instillation to pregnant C57BL/6 female mice with PM2.5 suspension or normal saline. We found that prenatal exposure to PM2.5 of a mouse model reduced body weight in adult male offspring after 6 weeks old. Histological analysis showed that the adipocyte size was significantly reduced in epididymal adipose tissue (eWAT) in male offspring, but not in brown adipose tissue. The expression levels of genes related to fatty acid synthesis (ACC1, ACSL1) and oxidation (PPARα) in eWAT were also significantly decreased. In addition, downregulation of pro-inflammatory cytokines (TNFα, IL-1β, IL-6) was also observed. Lipidomics analysis of eWAT demonstrated that prenatal exposure of PM2.5 reduced lysophosphatidylcholines (LPC), phosphatidylcholines (PC), phosphatidylethanolamines (PE), sphingomyelins (SM), and ceramides (Cer), indicating that metabolic pathways, including SM-Cer signaling and glycerophospholipids remodeling, were disrupted. In summary, prenatal exposure to PM2.5 was associated with the dysregulations in lipid metabolism of eWAT and pro-inflammatory response in male offspring.
Collapse
Affiliation(s)
- Peisi Xie
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Chao Zhao
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Wei Huang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Ting Yong
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Arthur C K Chung
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Kaiwu He
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xiangfeng Chen
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, SAR, China; HKBU Institute for Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
106
|
Alcalá M, Calderon-Dominguez M, Serra D, Herrero L, Viana M. Mechanisms of Impaired Brown Adipose Tissue Recruitment in Obesity. Front Physiol 2019; 10:94. [PMID: 30814954 PMCID: PMC6381290 DOI: 10.3389/fphys.2019.00094] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
Brown adipose tissue (BAT) dissipates energy to produce heat. Thus, it has the potential to regulate body temperature by thermogenesis. For the last decade, BAT has been in the spotlight due to its rediscovery in adult humans. This is evidenced by over a hundred clinical trials that are currently registered to target BAT as a therapeutic tool in the treatment of metabolic diseases, such as obesity or diabetes. The goal of most of these trials is to activate the BAT thermogenic program via several approaches such as adrenergic stimulation, natriuretic peptides, retinoids, capsinoids, thyroid hormones, or glucocorticoids. However, the impact of BAT activation on total body energy consumption and the potential effect on weight loss is still limited. Other studies have focused on increasing the mass of thermogenic BAT. This can be relevant in obesity, where the activity and abundance of BAT have been shown to be drastically reduced. The aim of this review is to describe pathological processes associated with obesity that may influence the correct differentiation of BAT, such as catecholamine resistance, inflammation, oxidative stress, and endoplasmic reticulum stress. This will shed light on the thermogenic potential of BAT as a therapeutic approach to target obesity-induced metabolic diseases.
Collapse
Affiliation(s)
- Martín Alcalá
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María Calderon-Dominguez
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Viana
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
107
|
Ro SH, Jang Y, Bae J, Kim IM, Schaecher C, Shomo ZD. Autophagy in Adipocyte Browning: Emerging Drug Target for Intervention in Obesity. Front Physiol 2019; 10:22. [PMID: 30745879 PMCID: PMC6360992 DOI: 10.3389/fphys.2019.00022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/10/2019] [Indexed: 12/27/2022] Open
Abstract
Autophagy, lipophagy, and mitophagy are considered to be the major recycling processes for protein aggregates, excess fat, and damaged mitochondria in adipose tissues in response to nutrient status-associated stress, oxidative stress, and genotoxic stress in the human body. Obesity with increased body weight is often associated with white adipose tissue (WAT) hypertrophy and hyperplasia and/or beige/brown adipose tissue atrophy and aplasia, which significantly contribute to the imbalance in lipid metabolism, adipocytokine secretion, free fatty acid release, and mitochondria function. In recent studies, hyperactive autophagy in WAT was observed in obese and diabetic patients, and inhibition of adipose autophagy through targeted deletion of autophagy genes in mice improved anti-obesity phenotypes. In addition, active mitochondria clearance through activation of autophagy was required for beige/brown fat whitening – that is, conversion to white fat. However, inhibition of autophagy seemed detrimental in hypermetabolic conditions such as hepatic steatosis, atherosclerosis, thermal injury, sepsis, and cachexia through an increase in free fatty acid and glycerol release from WAT. The emerging concept of white fat browning–conversion to beige/brown fat–has been controversial in its anti-obesity effect through facilitation of weight loss and improving metabolic health. Thus, proper regulation of autophagy activity fit to an individual metabolic profile is necessary to ensure balance in adipose tissue metabolism and function, and to further prevent metabolic disorders such as obesity and diabetes. In this review, we summarize the effect of autophagy in adipose tissue browning in the context of obesity prevention and its potential as a promising target for the development of anti-obesity drugs.
Collapse
Affiliation(s)
- Seung-Hyun Ro
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Yura Jang
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jiyoung Bae
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Isaac M Kim
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Cameron Schaecher
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States.,College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Zachery D Shomo
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
108
|
Yao X, Salingova B, Dani C. Brown-Like Adipocyte Progenitors Derived from Human iPS Cells: A New Tool for Anti-obesity Drug Discovery and Cell-Based Therapy? Handb Exp Pharmacol 2019; 251:97-105. [PMID: 29633179 DOI: 10.1007/164_2018_115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Alternative strategies are urgently required to fight obesity and associated metabolic disorders including diabetes and cardiovascular diseases. Brown and brown-like adipocytes (BAs) store fat, but in contrast to white adipocytes, activated BAs are equipped to dissipate energy stored. Therefore, BAs represent promising cell targets to counteract obesity. However, the scarcity of BAs in adults is a major limitation for a BA-based therapy of obesity, and the notion to increase the BA mass by transplanting BA progenitors (BAPs) in obese patients recently emerged. The next challenge is to identify an abundant and reliable source of BAPs. In this chapter, we describe the capacity of human-induced pluripotent stem cells (hiPSCs) to generate BAPs able to differentiate at a high efficiency with no gene transfer. This cell model represents an unlimited source of human BAPs that in a near future may be a suitable tool for both therapeutic transplantation and for the discovery of novel efficient and safe anti-obesity drugs. The generation of a relevant cell model, such as hiPSC-BAs in 3D adipospheres enriched with macrophages and endothelial cells to better mimic the microenvironment within the adipose tissue, will be the next critical step.
Collapse
Affiliation(s)
- Xi Yao
- Faculté de Médecine, Université Nice Sophia Antipolis, iBV, UMR CNRS/INSERM, Nice, Cedex 2, France
| | - Barbara Salingova
- Faculté de Médecine, Université Nice Sophia Antipolis, iBV, UMR CNRS/INSERM, Nice, Cedex 2, France
| | - Christian Dani
- Faculté de Médecine, Université Nice Sophia Antipolis, iBV, UMR CNRS/INSERM, Nice, Cedex 2, France.
| |
Collapse
|
109
|
Takagi M, Kimura K, Nakashima KI, Hirai T, Inoue M. Induction of beige adipocytes by naturally occurring β3-adrenoceptor agonist p-synephrine. Eur J Pharmacol 2018; 836:67-74. [DOI: 10.1016/j.ejphar.2018.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/31/2022]
|
110
|
BATLAS: Deconvoluting Brown Adipose Tissue. Cell Rep 2018; 25:784-797.e4. [DOI: 10.1016/j.celrep.2018.09.044] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 01/18/2023] Open
|
111
|
García MDC, Pazos P, Lima L, Diéguez C. Regulation of Energy Expenditure and Brown/Beige Thermogenic Activity by Interleukins: New Roles for Old Actors. Int J Mol Sci 2018; 19:E2569. [PMID: 30158466 PMCID: PMC6164446 DOI: 10.3390/ijms19092569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/25/2018] [Indexed: 12/16/2022] Open
Abstract
Obesity rates and the burden of metabolic associated diseases are escalating worldwide Energy burning brown and inducible beige adipocytes in human adipose tissues (ATs) have attracted considerable attention due to their therapeutic potential to counteract the deleterious metabolic effects of nutritional overload and overweight. Recent research has highlighted the relevance of resident and recruited ATs immune cell populations and their signalling mediators, cytokines, as modulators of the thermogenic activity of brown and beige ATs. In this review, we first provide an overview of the developmental, cellular and functional heterogeneity of the AT organ, as well as reported molecular switches of its heat-producing machinery. We also discuss the key contribution of various interleukins signalling pathways to energy and metabolic homeostasis and their roles in the biogenesis and function of brown and beige adipocytes. Besides local actions, attention is also drawn to their influence in the central nervous system (CNS) networks governing energy expenditure.
Collapse
Affiliation(s)
- María Del Carmen García
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III (ISCIII, Ministerio de Economía y Competitividad (MINECO)), C/Monforte de Lemos 3-5, Pabellón 11. Planta 0, 28029 Madrid, Spain.
| | - Patricia Pazos
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III (ISCIII, Ministerio de Economía y Competitividad (MINECO)), C/Monforte de Lemos 3-5, Pabellón 11. Planta 0, 28029 Madrid, Spain.
| | - Luis Lima
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
| | - Carlos Diéguez
- Department of Physiology/Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain.
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III (ISCIII, Ministerio de Economía y Competitividad (MINECO)), C/Monforte de Lemos 3-5, Pabellón 11. Planta 0, 28029 Madrid, Spain.
| |
Collapse
|
112
|
Zhao C, Zeng Z, Qazvini NT, Yu X, Zhang R, Yan S, Shu Y, Zhu Y, Duan C, Bishop E, Lei J, Zhang W, Yang C, Wu K, Wu Y, An L, Huang S, Ji X, Gong C, Yuan C, Zhang L, Liu W, Huang B, Feng Y, Zhang B, Dai Z, Shen Y, Wang X, Luo W, Oliveira L, Athiviraham A, Lee MJ, Wolf JM, Ameer GA, Reid RR, He TC, Huang W. Thermoresponsive Citrate-Based Graphene Oxide Scaffold Enhances Bone Regeneration from BMP9-Stimulated Adipose-Derived Mesenchymal Stem Cells. ACS Biomater Sci Eng 2018; 4:2943-2955. [PMID: 30906855 PMCID: PMC6425978 DOI: 10.1021/acsbiomaterials.8b00179] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/07/2018] [Indexed: 02/06/2023]
Abstract
Effective bone tissue engineering is important to overcome the unmet clinical challenges as more than 1.6 million bone grafts are done annually in the United States. Successful bone tissue engineering needs minimally three critical constituents: osteoprogenitor cells, osteogenic factors, and osteoinductive/osteoconductive scaffolds. Osteogenic progenitors are derived from multipotent mesenchymal stem cells (MSCs), which can be prepared from numerous tissue sources, including adipose tissue. We previously showed that BMP9 is the most osteogenic BMP and induces robust bone formation of immortalized mouse adipose-derived MSCs entrapped in a citrate-based thermoresponsive hydrogel referred to as PPCNg. As graphene and its derivatives emerge as promising biomaterials, here we develop a novel thermosensitive and injectable hybrid material by combining graphene oxide (GO) with PPCNg (designated as GO-P) and characterize its ability to promote bone formation. We demonstrate that the thermoresponsive behavior of the hybrid material is maintained while effectively supporting MSC survival and proliferation. Furthermore, GO-P induces early bone-forming marker alkaline phosphatase (ALP) and potentiates BMP9-induced expression of osteogenic regulators and bone markers as well as angiogenic factor VEGF in MSCs. In vivo studies show BMP9-transduced MSCs entrapped in the GO-P scaffold form well-mineralized and highly vascularized trabecular bone. Thus, these results indicate that GO-P hybrid material may function as a new biocompatible, injectable scaffold with osteoinductive and osteoconductive activities for bone regeneration.
Collapse
Affiliation(s)
- Chen Zhao
- Departments
of Orthopedic Surgery, Nephrology, Cardiology, Clinical Laboratory
Medicine, and Breast Surgery, The First
Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Zongyue Zeng
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Nader Taheri Qazvini
- Institute
for Molecular Engineering, The University
of Chicago, 5640 South
Ellis Avenue, Chicago, Illinois 60637, United States
| | - Xinyi Yu
- Departments
of Orthopedic Surgery, Nephrology, Cardiology, Clinical Laboratory
Medicine, and Breast Surgery, The First
Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Ruyi Zhang
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Shujuan Yan
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Yi Shu
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Yunxiao Zhu
- Department
of Biomedical Engineering, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
- Center for Advanced Regenerative Engineering (CARE), 2145 Sheridan Road, Evanston, IL 60208, United
States
| | - Chongwen Duan
- Department
of Biomedical Engineering, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
| | - Elliot Bishop
- Department
of Surgery, Laboratory of Craniofacial Biology and Development, Section
of Plastic Surgery, The University of Chicago
Medical Center, 5841
South Maryland Avenue MC6035, Chicago, Illinois 60637, United States
| | - Jiayan Lei
- Departments
of Orthopedic Surgery, Nephrology, Cardiology, Clinical Laboratory
Medicine, and Breast Surgery, The First
Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Wenwen Zhang
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Department
of Laboratory Medicine and Clinical Diagnostics, The Affiliated University-Town Hospital of Chongqing Medical University, 55 Daxuecheng Zhonglu, Chongqing 401331, China
| | - Chao Yang
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Ke Wu
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Ying Wu
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Department
of Immunology and Microbiology, Beijing
University of Chinese Medicine, 11 N. Third Ring Road E., Beijing 100029, China
| | - Liping An
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Key
Laboratory of Orthopaedic Surgery of Gansu Province and the Department
of Orthopaedic Surgery, The Second Hospital
of Lanzhou University, 82 Cuiyingmen, Lanzhou 730030, China
| | - Shifeng Huang
- Departments
of Orthopedic Surgery, Nephrology, Cardiology, Clinical Laboratory
Medicine, and Breast Surgery, The First
Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Xiaojuan Ji
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Cheng Gong
- Department of General Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Chengfu Yuan
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Department
of Biochemistry and Molecular Biology, China
Three Gorges University School of Medicine, 8 Daxue Road, Yichang 443002, China
| | - Linghuan Zhang
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Wei Liu
- Departments
of Orthopedic Surgery, Nephrology, Cardiology, Clinical Laboratory
Medicine, and Breast Surgery, The First
Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Bo Huang
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Yixiao Feng
- Departments
of Orthopedic Surgery, Nephrology, Cardiology, Clinical Laboratory
Medicine, and Breast Surgery, The First
Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Bo Zhang
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Key
Laboratory of Orthopaedic Surgery of Gansu Province and the Department
of Orthopaedic Surgery, The Second Hospital
of Lanzhou University, 82 Cuiyingmen, Lanzhou 730030, China
| | - Zhengyu Dai
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Department
of Orthopaedic Surgery, Chongqing Hospital
of Traditional Chinese Medicine, 35 Jianxin East Road, Chongqing 400021, China
| | - Yi Shen
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Department
of Orthopaedic Surgery, Xiangya Second Hospital
of Central South University, 139 Renmin Road, Changsha 410011, China
| | - Xi Wang
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Wenping Luo
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
| | - Leonardo Oliveira
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Aravind Athiviraham
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Michael J. Lee
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Jennifer Moriatis Wolf
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
| | - Guillermo A. Ameer
- Department
of Biomedical Engineering, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
- Department
of Surgery, Feinberg School of Medicine, Northwestern University, 420 East Superior Street, Chicago, Illinois 60616, United
States
- Center for Advanced Regenerative Engineering (CARE), 2145 Sheridan Road, Evanston, IL 60208, United
States
| | - Russell R. Reid
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Department
of Surgery, Laboratory of Craniofacial Biology and Development, Section
of Plastic Surgery, The University of Chicago
Medical Center, 5841
South Maryland Avenue MC6035, Chicago, Illinois 60637, United States
- Center for Advanced Regenerative Engineering (CARE), 2145 Sheridan Road, Evanston, IL 60208, United
States
| | - Tong-Chuan He
- Molecular
Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation
Medicine, The University of Chicago Medical
Center, 5841 South Maryland
Avenue MC 3079, Chicago, Illinois 60637, United
States
- Ministry
of Education Key Laboratory of Diagnostic Medicine and School of Laboratory
Medicine, The Affiliated Hospitals of Chongqing
Medical University, 1
Medical College Road, Chongqing 400016, China
- Center for Advanced Regenerative Engineering (CARE), 2145 Sheridan Road, Evanston, IL 60208, United
States
| | - Wei Huang
- Departments
of Orthopedic Surgery, Nephrology, Cardiology, Clinical Laboratory
Medicine, and Breast Surgery, The First
Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing 400016, China
| |
Collapse
|
113
|
|
114
|
Ramirez AK, Lynes MD, Shamsi F, Xue R, Tseng YH, Kahn CR, Kasif S, Dreyfuss JM. Integrating Extracellular Flux Measurements and Genome-Scale Modeling Reveals Differences between Brown and White Adipocytes. Cell Rep 2018; 21:3040-3048. [PMID: 29241534 DOI: 10.1016/j.celrep.2017.11.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 10/06/2017] [Accepted: 11/17/2017] [Indexed: 12/13/2022] Open
Abstract
White adipocytes are specialized for energy storage, whereas brown adipocytes are specialized for energy expenditure. Explicating this difference can help identify therapeutic targets for obesity. A common tool to assess metabolic differences between such cells is the Seahorse Extracellular Flux (XF) Analyzer, which measures oxygen consumption and media acidification in the presence of different substrates and perturbagens. Here, we integrate the Analyzer's metabolic profile from human white and brown adipocytes with a genome-scale metabolic model to predict flux differences across the metabolic map. Predictions matched experimental data for the metabolite 4-aminobutyrate, the protein ABAT, and the fluxes for glucose, glutamine, and palmitate. We also uncovered a difference in how adipocytes dispose of nitrogenous waste, with brown adipocytes secreting less ammonia and more urea than white adipocytes. Thus, the method and software we developed allow for broader metabolic phenotyping and provide a distinct approach to uncovering metabolic differences.
Collapse
Affiliation(s)
- Alfred K Ramirez
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Matthew D Lynes
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Farnaz Shamsi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ruidan Xue
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yu-Hua Tseng
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Simon Kasif
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Graduate Program in Bioinformatics, Boston University, Boston, MA 02215, USA.
| | - Jonathan M Dreyfuss
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Bioinformatics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
115
|
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
|
116
|
Schwalie PC, Dong H, Zachara M, Russeil J, Alpern D, Akchiche N, Caprara C, Sun W, Schlaudraff KU, Soldati G, Wolfrum C, Deplancke B. A stromal cell population that inhibits adipogenesis in mammalian fat depots. Nature 2018; 559:103-108. [PMID: 29925944 DOI: 10.1038/s41586-018-0226-8] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/16/2018] [Indexed: 02/07/2023]
Abstract
Adipocyte development and differentiation have an important role in the aetiology of obesity and its co-morbidities1,2. Although multiple studies have investigated the adipogenic stem and precursor cells that give rise to mature adipocytes3-14, our understanding of their in vivo origin and properties is incomplete2,15,16. This is partially due to the highly heterogeneous and unstructured nature of adipose tissue depots17, which has proven difficult to molecularly dissect using classical approaches such as fluorescence-activated cell sorting and Cre-lox lines based on candidate marker genes16,18. Here, using the resolving power of single-cell transcriptomics19 in a mouse model, we reveal distinct subpopulations of adipose stem and precursor cells in the stromal vascular fraction of subcutaneous adipose tissue. We identify one of these subpopulations as CD142+ adipogenesis-regulatory cells, which can suppress adipocyte formation in vivo and in vitro in a paracrine manner. We show that adipogenesis-regulatory cells are refractory to adipogenesis and that they are functionally conserved in humans. Our findings point to a potentially critical role for adipogenesis-regulatory cells in modulating adipose tissue plasticity, which is linked to metabolic control, differential insulin sensitivity and type 2 diabetes.
Collapse
Affiliation(s)
- Petra C Schwalie
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Hua Dong
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland
| | - Magda Zachara
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Julie Russeil
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nassila Akchiche
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland
| | | | - Wenfei Sun
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland
| | | | | | - Christian Wolfrum
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland.
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
117
|
Browning of white adipose tissue induced by the ß3 agonist CL-316,243 after local and systemic treatment - PK-PD relationship. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2972-2982. [PMID: 29902549 DOI: 10.1016/j.bbadis.2018.06.007] [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: 03/05/2018] [Revised: 05/20/2018] [Accepted: 06/09/2018] [Indexed: 12/12/2022]
Abstract
Transformation of white adipose tissue (WAT) to a brown adipose tissue-like (BAT-like) phenotype has emerged as an attractive approach against obesity e.g. using g ß3 adrenergic receptor agonists. These could however, produce side-effects following systemic exposure. The present study explored the possibility of local use of CL-316,243 - a selective ß3 agonist - to circumvent this problem. Rats treated s.c. for 2 weeks (0.3 and 1 mg/kg) showed decreased inguinal fat pad (IFP) weight/volume, increased UCP-1 staining and expressed BAT-like features in H&E stained micrographs. Interscapular BAT increased in weight/volume. In contrast, local treatment into the IFP was not efficacious in terms of weight/volume, despite slight increases in UCP-1 staining and changes in histological features. After local treatment, the exposure of the IFP was lower than after systemic treatment. In turn higher local doses (0.5 and 5 mg/ml) were then tested which produced a strong trend for decreased volume of the IFP, a significant increase in UCP-1 staining, and also a decrease in adipocytes size but increased number. However, after this treatment the systemic exposure was in the same range as following systemic treatment. In conclusion, we saw no evidence for the possibility of converting inguinal WAT to a BAT-phenotype solely through local activation of ß3 receptors. This is in concert with our in vitro experiments which detected direct effects of PPARγ agonists at the gene/protein expression and functional level, but were unable to detect any effect of CL-316,243.
Collapse
|
118
|
Singh AJ, Chang CN, Ma HY, Ramsey SA, Filtz TM, Kioussi C. FACS-Seq analysis of Pax3-derived cells identifies non-myogenic lineages in the embryonic forelimb. Sci Rep 2018; 8:7670. [PMID: 29769607 PMCID: PMC5956100 DOI: 10.1038/s41598-018-25998-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/01/2018] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle in the forelimb develops during embryonic and fetal development and perinatally. While much is known regarding the molecules involved in forelimb myogenesis, little is known about the specific mechanisms and interactions. Migrating skeletal muscle precursor cells express Pax3 as they migrate into the forelimb from the dermomyotome. To compare gene expression profiles of the same cell population over time, we isolated lineage-traced Pax3+ cells (Pax3EGFP) from forelimbs at different embryonic days. We performed whole transcriptome profiling via RNA-Seq of Pax3+ cells to construct gene networks involved in different stages of embryonic and fetal development. With this, we identified genes involved in the skeletal, muscular, vascular, nervous and immune systems. Expression of genes related to the immune, skeletal and vascular systems showed prominent increases over time, suggesting a non-skeletal myogenic context of Pax3-derived cells. Using co-expression analysis, we observed an immune-related gene subnetwork active during fetal myogenesis, further implying that Pax3-derived cells are not a strictly myogenic lineage, and are involved in patterning and three-dimensional formation of the forelimb through multiple systems.
Collapse
Affiliation(s)
- Arun J Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA.,Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Hsiao-Yen Ma
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Stephen A Ramsey
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, 97331, USA.,School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Theresa M Filtz
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, 97331, USA.
| |
Collapse
|
119
|
Singh AM, Dalton S. What Can 'Brown-ing' Do For You? Trends Endocrinol Metab 2018; 29:349-359. [PMID: 29606342 PMCID: PMC5937921 DOI: 10.1016/j.tem.2018.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/31/2022]
Abstract
Human stem cell-based models of thermogenic adipocytes provide an opportunity for the establishment of new therapeutics, modeling of disease mechanisms, and understanding of development. Pluripotent stem cells, adipose-derived stem cells/preadipocytes, and programming-reprogramming-based approaches have been used to develop cell-based platforms for drug screening and transplantable therapeutics in the metabolic disease arena. Here we provide a detailed overview of these approaches, the latest advances in this field, and the opportunities and shortcomings they present. Moreover, we comment on how stem-cell-based platforms can be best utilized in the future for the treatment and understanding of metabolic diseases, including type 2 diabetes and associated medical issues such as obesity.
Collapse
Affiliation(s)
- Amar M Singh
- Center for Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA
| | - Stephen Dalton
- Center for Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Georgia, 325 Riverbend Road, Athens, GA 30602, USA.
| |
Collapse
|
120
|
Li Z, Hardij J, Bagchi DP, Scheller EL, MacDougald OA. Development, regulation, metabolism and function of bone marrow adipose tissues. Bone 2018; 110:134-140. [PMID: 29343445 PMCID: PMC6277028 DOI: 10.1016/j.bone.2018.01.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 01/08/2018] [Indexed: 12/29/2022]
Abstract
Most adipocytes exist in discrete depots throughout the body, notably in well-defined white and brown adipose tissues. However, adipocytes also reside within specialized niches, of which the most abundant is within bone marrow. Whereas bone marrow adipose tissue (BMAT) shares many properties in common with white adipose tissue, the distinct functions of BMAT are reflected by its development, regulation, protein secretion, and lipid composition. In addition to its potential role as a local energy reservoir, BMAT also secretes proteins, including adiponectin, RANK ligand, dipeptidyl peptidase-4, and stem cell factor, which contribute to local marrow niche functions and which may also influence global metabolism. The characteristics of BMAT are also distinct depending on whether marrow adipocytes are contained within yellow or red marrow, as these can be thought of as 'constitutive' and 'regulated', respectively. The rBMAT for instance can be expanded or depleted by myriad factors, including age, nutrition, endocrine status and pharmaceuticals. Herein we review the site specificity, age-related development, regulation and metabolic characteristics of BMAT under various metabolic conditions, including the functional interactions with bone and hematopoietic cells.
Collapse
Affiliation(s)
- Ziru Li
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Julie Hardij
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Devika P Bagchi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Erica L Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, MO, United States
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
121
|
Suchacki KJ, Cawthorn WP. Molecular Interaction of Bone Marrow Adipose Tissue with Energy Metabolism. CURRENT MOLECULAR BIOLOGY REPORTS 2018; 4:41-49. [PMID: 29888168 PMCID: PMC5976678 DOI: 10.1007/s40610-018-0096-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The last decade has seen a resurgence in the study of bone marrow adipose tissue (BMAT) across diverse fields such as metabolism, haematopoiesis, skeletal biology and cancer. Herein, we review the most recent developments of BMAT research in both humans and rodents, including the distinct nature of BMAT; the autocrine, paracrine and endocrine interactions between BMAT and various tissues, both in physiological and pathological scenarios; how these interactions might impact energy metabolism; and the most recent technological advances to quantify BMAT. RECENT FINDINGS Though still dwarfed by research into white and brown adipose tissues, BMAT is now recognised as endocrine organ and is attracting increasing attention from biomedical researchers around the globe. SUMMARY We are beginning to learn the importance of BMAT both within and beyond the bone, allowing us to better appreciate the role of BMAT in normal physiology and disease.
Collapse
Affiliation(s)
- Karla J. Suchacki
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ UK
| | - William P. Cawthorn
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ UK
| |
Collapse
|
122
|
Aldiss P, Betts J, Sale C, Pope M, Budge H, Symonds ME. Exercise-induced 'browning' of adipose tissues. Metabolism 2018; 81:63-70. [PMID: 29155135 PMCID: PMC5893183 DOI: 10.1016/j.metabol.2017.11.009] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/30/2017] [Accepted: 11/13/2017] [Indexed: 12/18/2022]
Abstract
Global rates of obesity continue to rise and are necessarily the consequence of a long-term imbalance between energy intake and energy expenditure. This is the result of an expansion of adipose tissue due to both the hypertrophy of existing adipocytes and hyperplasia of adipocyte pre-cursors. Exercise elicits numerous physiological benefits on adipose tissue, which are likely to contribute to the associated cardiometabolic benefits. More recently it has been demonstrated that exercise, through a range of mechanisms, induces a phenotypic switch in adipose tissue from energy storing white adipocytes to thermogenic beige adipocytes. This has generated the hypothesis that the process of adipocyte 'browning' may partially underlie the improved cardiometabolic health in physically active populations. Interestingly, 'browning' also occurs in response to various stressors and could represent an adaptive response. In the context of exercise, it is not clear whether the appearance of beige adipocytes is metabolically beneficial or whether they occur as a transient adaptive process to exercise-induced stresses. The present review discusses the various mechanisms (e.g. fatty acid oxidation during exercise, decreased thermal insulation, stressors and angiogenesis) by which the exercise-induced 'browning' process may occur.
Collapse
Affiliation(s)
- Peter Aldiss
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham NG7 2UH, UK
| | - James Betts
- Department for Health, University of Bath, Bath, BA2 7AY, UK
| | - Craig Sale
- Musculoskeletal Physiology Research Group, Sport, Health and Performance Enhancement Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Mark Pope
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham NG7 2UH, UK
| | - Helen Budge
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham NG7 2UH, UK
| | - Michael E Symonds
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, University of Nottingham, Nottingham NG7 2UH, UK; Nottingham Digestive Disease Centre and Biomedical Research Centre School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH.
| |
Collapse
|
123
|
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: 125] [Impact Index Per Article: 17.9] [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
|
124
|
Guaita-Esteruelas S, Gumà J, Masana L, Borràs J. The peritumoural adipose tissue microenvironment and cancer. The roles of fatty acid binding protein 4 and fatty acid binding protein 5. Mol Cell Endocrinol 2018; 462:107-118. [PMID: 28163102 DOI: 10.1016/j.mce.2017.02.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/11/2017] [Accepted: 02/01/2017] [Indexed: 02/08/2023]
Abstract
The adipose tissue microenvironment plays a key role in tumour initiation and progression because it provides fatty acids and adipokines to tumour cells. The fatty acid-binding protein (FABP) family is a group of small proteins that act as intracellular fatty acid transporters. Adipose-derived FABPs include FABP4 and FABP5. Both have an important role in lipid-related metabolic processes and overexpressed in many cancers, such as breast, prostate, colorectal and ovarian. Moreover, their expression in peritumoural adipose tissue is deregulated, and their circulating levels are upregulated in some tumours. In this review, we discuss the role of the peritumoural adipose tissue and the related adipokines FABP4 and FABP5 in cancer initiation and progression and the possible pathways implicated in these processes.
Collapse
Affiliation(s)
- S Guaita-Esteruelas
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Centre d'R+D+I en Nutrició i Salut, Avda. de la Universitat, 43204 Reus, Spain; Research Unit on Lipids and Atherosclerosis, Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Universitat Rovira i Virgili, Sant Llorenç, 21 43201 Reus, Spain; Institut d'Oncologia de la Catalunya Sud (IOCS), Hospital Universitari Sant Joan de Reus, IISPV, Universitat Rovira i Virgili, Av. del Dr, Josep Laporte, 2, 43204 Reus, Spain.
| | - J Gumà
- Institut d'Oncologia de la Catalunya Sud (IOCS), Hospital Universitari Sant Joan de Reus, IISPV, Universitat Rovira i Virgili, Av. del Dr, Josep Laporte, 2, 43204 Reus, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain
| | - L Masana
- Research Unit on Lipids and Atherosclerosis, Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Universitat Rovira i Virgili, Sant Llorenç, 21 43201 Reus, Spain
| | - J Borràs
- Institut d'Oncologia de la Catalunya Sud (IOCS), Hospital Universitari Sant Joan de Reus, IISPV, Universitat Rovira i Virgili, Av. del Dr, Josep Laporte, 2, 43204 Reus, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain
| |
Collapse
|
125
|
Gao Z, Daquinag AC, Su F, Snyder B, Kolonin MG. PDGFRα/PDGFRβ signaling balance modulates progenitor cell differentiation into white and beige adipocytes. Development 2018; 145:dev.155861. [PMID: 29158445 DOI: 10.1242/dev.155861] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/13/2017] [Indexed: 12/19/2022]
Abstract
The relative abundance of thermogenic beige adipocytes and lipid-storing white adipocytes in adipose tissue underlie its metabolic activity. The roles of adipocyte progenitor cells, which express PDGFRα or PDGFRβ, in adipose tissue function have remained unclear. Here, by defining the developmental timing of PDGFRα and PDGFRβ expression in mouse subcutaneous and visceral adipose depots, we uncover depot specificity of pre-adipocyte delineation. We demonstrate that PDGFRα expression precedes PDGFRβ expression in all subcutaneous but in only a fraction of visceral adipose stromal cells. We show that high-fat diet feeding or thermoneutrality in early postnatal development can induce PDGFRβ+ lineage recruitment to generate white adipocytes. In contrast, the contribution of PDGFRβ+ lineage to beige adipocytes is minimal. We provide evidence that human adipose tissue also contains distinct progenitor populations differentiating into beige or white adipocytes, depending on PDGFRβ expression. Based on PDGFRα or PDGFRβ deletion and ectopic expression experiments, we conclude that the PDGFRα/PDGFRβ signaling balance determines progenitor commitment to beige (PDGFRα) or white (PDGFRβ) adipogenesis. Our study suggests that adipocyte lineage specification and metabolism can be modulated through PDGFR signaling.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Alexes C Daquinag
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Fei Su
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Brad Snyder
- Department of Surgery, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
126
|
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
|
127
|
Frank AP, Palmer BF, Clegg DJ. Do estrogens enhance activation of brown and beiging of adipose tissues? Physiol Behav 2017; 187:24-31. [PMID: 28988965 DOI: 10.1016/j.physbeh.2017.09.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/30/2017] [Accepted: 09/30/2017] [Indexed: 01/05/2023]
Abstract
Obesity and its associated co-morbidities are worldwide public health concerns. Obesity is characterized by excessive adipose tissue accumulation; however, it is important to recognize that human and rodent adipose tissues are made up of several distinct adipose tissue sub-types. White adipose tissue (WAT) is considered the prototypical fat cell, due to its capacity and capability to store large amounts of lipid. In contrast, brown adipose tissue (BAT) oxidizes substrates to generate heat. BAT contains more mitochondria than WAT and express uncoupling protein-1 (UCP1), which mediates BAT thermogenesis. A third sub-type of adipose tissue, Brown-in-white (BRITE)/beige adipocytes arise from WAT upon adrenergic stimulation and resembles BAT functionally. The energy burning feature of BAT/beige cells, combined with evidence of an inverse-correlation between BAT/beige adipose tissue and obesity have given rise to the hypothesis that obesity may be linked to BAT/beige 'malfunction'. Females have more BAT and perhaps an enhanced capacity to beige their adipose tissue when compared to males. Multiple signal pathways are capable of activating BAT thermogenesis and beiging of WAT; here, we discuss the potential role of estrogens in enhancing and mediating these factors to enhance adipose tissue thermogenesis.
Collapse
Affiliation(s)
- Aaron P Frank
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Deborah J Clegg
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
128
|
Bharath LP, Ip BC, Nikolajczyk BS. Adaptive Immunity and Metabolic Health: Harmony Becomes Dissonant in Obesity and Aging. Compr Physiol 2017; 7:1307-1337. [PMID: 28915326 DOI: 10.1002/cphy.c160042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipose tissue (AT) is the primary energy reservoir organ, and thereby plays a critical role in energy homeostasis and regulation of metabolism. AT expands in response to chronic overnutrition or aging and becomes a major source of inflammation that has marked influence on systemic metabolism. The chronic, sterile inflammation that occurs in the AT during the development of obesity or in aging contributes to onset of devastating diseases such as insulin resistance, diabetes, and cardiovascular pathologies. Numerous studies have shown that inflammation in the visceral AT of humans and animals is a critical trigger for the development of metabolic syndrome. This work underscores the well-supported conclusion that the inflammatory immune response and metabolic pathways in the AT are tightly interwoven by multiple layers of relatively conserved mechanisms. During the development of diet-induced obesity or age-associated adiposity, cells of the innate and the adaptive immune systems infiltrate and proliferate in the AT. Macrophages, which dominate AT-associated immune cells in mouse models of obesity, but are less dominant in obese people, have been studied extensively. However, cells of the adaptive immune system, including T cells and B cells, contribute significantly to AT inflammation, perhaps more in humans than in mice. Lymphocytes regulate recruitment of innate immune cells into AT, and produce cytokines that influence the helpful-to-harmful inflammatory balance that, in turn, regulates organismal metabolism. This review describes inflammation, or more precisely, metabolic inflammation (metaflammation) with an eye toward the AT and the roles lymphocytes play in regulation of systemic metabolism during obesity and aging. © 2017 American Physiological Society. Compr Physiol 7:1307-1337, 2017.
Collapse
Affiliation(s)
- Leena P Bharath
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Blanche C Ip
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Molecular Pharmacology, Physiology and Biotechnology, Center of Biomedical Engineering, Brown University, Providence, Rhode Island, USA
| | | |
Collapse
|
129
|
Pervin S, Singh V, Tucker A, Collazo J, Singh R. Modulation of transforming growth factor-β/follistatin signaling and white adipose browning: therapeutic implications for obesity related disorders. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0036/hmbci-2017-0036.xml. [PMID: 28888087 DOI: 10.1515/hmbci-2017-0036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/17/2017] [Indexed: 12/15/2022]
Abstract
Obesity is a major risk factor for the development of diabetes, insulin resistance, dyslipidemia, cardiovascular disease and other related metabolic conditions. Obesity develops from perturbations in overall cellular bioenergetics when energy intake chronically exceeds total energy expenditure. Lifestyle interventions based on reducing total energy uptake and increasing activities including exercise have proved ineffective in the prevention and treatment of obesity because of poor adherence to such interventions for an extended period of time. Brown adipose tissue (BAT) has an extraordinary metabolic capacity to burn excess stored energy and holds great promise in combating obesity and related diseases. This unique ability to nullify the effects of extra energy intake of these specialized tissues has provided attractive perspectives for the therapeutic potential of BAT in humans. Browning of white adipose tissue by promoting the expression and activity of key mitochondrial uncoupling protein 1 (UCP1) represents an exciting new strategy to combat obesity via enhanced energy dissipation. Members of the transforming growth factor-beta (TGF-β) superfamily including myostatin and follistatin have recently been demonstrated to play a key role in regulating white adipose browning both in in-vitro and in-vivo animal models and thereby present attractive avenues for exploring the therapeutic potential for the treatment of obesity and related metabolic diseases.
Collapse
|
130
|
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
|
131
|
Stolarczyk E. Adipose tissue inflammation in obesity: a metabolic or immune response? Curr Opin Pharmacol 2017; 37:35-40. [PMID: 28843953 DOI: 10.1016/j.coph.2017.08.006] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/31/2017] [Accepted: 08/07/2017] [Indexed: 12/21/2022]
Abstract
Adipose tissue is not only a reservoir for energy, but also an immune organ. In the context of obesity, the development of insulin resistance is now recognised to be initiated by inflammation of the adipose tissue. However, the primary events triggering this inflammation are still unclear, as a complex combination of endocrine and immune factors act to regulate this adipose tissue microenvironment. Below we discuss the different factors involved and how they affect the biology of the adipose tissue in obesity.
Collapse
Affiliation(s)
- Emilie Stolarczyk
- Division of Diabetes, Endocrinology and Metabolism, Hammersmith Campus, Imperial College London, London, UK.
| |
Collapse
|
132
|
Ologun GO, Patel ZM, Rana NK, Trecartin A, Shen A, Trostle D, Bertsch D. Large Unilateral Adrenal Mass with Surrounding Brown Fat: A Case Report. Cureus 2017; 9:e1552. [PMID: 29021924 PMCID: PMC5633263 DOI: 10.7759/cureus.1552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Pheochromocytomas are rare tumors derived from chromaffin cells located in the adrenal and extra adrenal tissues. Pheochromocytomas are diagnosed biochemically and localized using different imaging modalities. The definitive management is surgical resection. Brown adipose tissues are normally present during fetal development, with regression over time. Brown adipose tissues are thermogenic and usually located in the neck, mediastinum, and retroperitoneum. Here, we report a case of a unilateral pheochromocytoma surrounded by brown fat. The abnormal stimulation of brown fat noted on positive emission tomography scan (PET) resolved after the pheochromocytoma was resected.
Collapse
Affiliation(s)
| | | | | | | | - Alice Shen
- General Surgery, Guthrie Clinic/Robert Packer Hospital
| | | | - David Bertsch
- Surgical Oncology, Guthrie Clinic/Robert Packer Hospital
| |
Collapse
|
133
|
Li L, Li B, Li M, Niu C, Wang G, Li T, Król E, Jin W, Speakman JR. Brown adipocytes can display a mammary basal myoepithelial cell phenotype in vivo. Mol Metab 2017; 6:1198-1211. [PMID: 29031720 PMCID: PMC5641686 DOI: 10.1016/j.molmet.2017.07.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 07/31/2017] [Indexed: 01/04/2023] Open
Abstract
Objective Previous work has suggested that white adipocytes may also show a mammary luminal secretory cell phenotype during lactation. The capacity of brown and beige/brite adipocytes to display a mammary cell phenotype and the levels at which they demonstrate such phenotypes in vivo is currently unknown. Methods To investigate the putative adipocyte origin of mammary gland cells, we performed genetic lineage-labeling experiments in BAT and the mammary glands. Results These studies indicated that the classic brown adipocytes (Ucp1+) and subcutaneous beige/brite adipocytes (Ucp1−/+) were found in the mammary gland during lactation, when they exhibited a mammary myoepithelial phenotype. Up to 2.5% of the anterior dorsal interscapular mammary myoepithelial cell population had a brown adipocyte origin with an adipose and myoepithelial gene signature during lactation. Eliminating these cells, along with all the brown adipocytes, significantly slowed offspring growth, potentially demonstrating their functional importance. Additionally, we showed mammary epithelial lineage Mmtv+ and Krt14+ cells expressed brown adipocyte markers after weaning, demonstrating that mammary gland cells can display an adipose phenotype. Conclusions The identification of a brown adipocyte origin of mammary myoepithelial cells provides a novel perspective on the interrelationships between adipocytes and mammary cells with implications for our understanding of obesity and breast cancer. Brown adipocytes can show a mammary myoepithelial cell phenotype in vivo. Myf5+/Ucp1+ myoepithelial cells express an adipose and myoepithelial signature. Mammary-derived epithelial cells can display adipose features after weaning.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoguo Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Min Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chaoqun Niu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guanlin Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ting Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Elżbieta Król
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK.
| |
Collapse
|
134
|
Mattiucci D, Maurizi G, Izzi V, Cenci L, Ciarlantini M, Mancini S, Mensà E, Pascarella R, Vivarelli M, Olivieri A, Leoni P, Poloni A. Bone marrow adipocytes support hematopoietic stem cell survival. J Cell Physiol 2017; 233:1500-1511. [PMID: 28574591 DOI: 10.1002/jcp.26037] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/01/2017] [Indexed: 12/23/2022]
Abstract
In bone marrow (BM), hematopoietic elements are mingled with adipocytes (BM-A), which are the most abundant stromal component in the niche. BM-A progressively increase with aging, eventually occupying up to 50% of BM cavities. In this work, the role played by BM-A was explored by studying primary human BM-A isolated from hip surgery patients at the molecular level, through microarray analysis, and at the functional level, by assessing their relationship with primary human hematopoietic stem cells (HSC) by the long-term culture initiating cell (LTC-IC) assay. Findings demonstrated that BM-A are capable of supporting HSC survival in the LTC-IC assay, since after 5 weeks of co-culture, HSC were still able to proliferate and differentiate. Furthermore, critical molecules such as C-X-C motif chemokine 12 (CXCL12), interleukin (IL)-8, colony-stimulating factor 3 (CSF3), and leukaemia inhibitory factor (LIF), were expressed at similar levels in BM-A and in primary human BM mesenchymal stromal cells (BM-MSC), whereas IL-3 was higher in BM-A. Interestingly, BM-A displayed a different gene expression profile compared with subcutaneous adipose tissue adipocytes (AT-A) collected from abdominal surgery patients, especially in terms of regulation of lipid metabolism, stemness genes, and white-to-brown differentiation pathways. Accordingly, analysis of the gene pathways involved in hematopoiesis regulation showed that BM-A are more closely related to BM-MSC than to AT-A. The present data suggest that BM-A play a supporting role in the hematopoietic niche and directly sustain HSC survival.
Collapse
Affiliation(s)
- Domenico Mattiucci
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Giulia Maurizi
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, Center for Cell-Matrix Research and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Lorenzo Cenci
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Marco Ciarlantini
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Stefania Mancini
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Emanuela Mensà
- Cardiology Unit, Italian National Research Center on Aging (INRCA-IRCCS), Experimental Models in Clinical Pathology, INRCA-IRCCS National Institute, Ancona, Italy
| | | | - Marco Vivarelli
- Hepatobiliary and Abdominal Transplantation Surgery, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Attilio Olivieri
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Pietro Leoni
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| | - Antonella Poloni
- Dipartimento Scienze Cliniche e Molecolari, Clinica di Ematologia, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
135
|
Chu DT, Tao Y, Son LH, Le DH. Cell source, differentiation, functional stimulation, and potential application of human thermogenic adipocytes in vitro. J Physiol Biochem 2017; 73:315-321. [PMID: 28612196 DOI: 10.1007/s13105-017-0567-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
Recent investigations have showed that the functional thermogenic adipocytes are present in both infants and adult humans. Accumulating evidence suggests that the coexistence of classical and inducible brown (brite) adipocytes in humans at adulthood and these adipocytes function to generate heat from energy resulting in reducing body fat and improving glucose metabolism. Human thermogenic adipocytes can be differentiated in vitro from stem cells, cell lines, or adipose stromal vascular fraction. Pre-activated human brite adipocytes in vitro can maintain their thermogenic function in normal or obese immunodeficient mice; therefore, they improve glucose homeostasis and reduce fat mass in obese animals. These key findings have opened a new door to use in vitro thermogenic adipocytes as a cell therapy to prevent obesity and related disorders. Thus, this paper intends to highlight our knowledge in aspects of in vitro human brite/brown adipocytes for the further studies.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Institute for Research and Development, Duy Tan University, 03 Quang Trung, Da Nang, Vietnam. .,Faculty of Biology, Hanoi National University of Education, Hanoi, Vietnam.
| | - Yang Tao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Vietnam
| | - Le Hoang Son
- VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Duc-Hau Le
- VINMEC Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam
| |
Collapse
|
136
|
Li JX, Cummins CL. Getting the Skinny on Follistatin and Fat. Endocrinology 2017; 158:1109-1112. [PMID: 28609834 DOI: 10.1210/en.2017-00223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/08/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Jia-Xu Li
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
137
|
den Broeder MJ, Moester MJB, Kamstra JH, Cenijn PH, Davidoiu V, Kamminga LM, Ariese F, de Boer JF, Legler J. Altered Adipogenesis in Zebrafish Larvae Following High Fat Diet and Chemical Exposure Is Visualised by Stimulated Raman Scattering Microscopy. Int J Mol Sci 2017; 18:E894. [PMID: 28441764 PMCID: PMC5412473 DOI: 10.3390/ijms18040894] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/08/2017] [Accepted: 04/18/2017] [Indexed: 02/08/2023] Open
Abstract
Early life stage exposure to environmental chemicals may play a role in obesity by altering adipogenesis; however, robust in vivo methods to quantify these effects are lacking. The goal of this study was to analyze the effects of developmental exposure to chemicals on adipogenesis in the zebrafish (Danio rerio). We used label-free Stimulated Raman Scattering (SRS) microscopy for the first time to image zebrafish adipogenesis at 15 days post fertilization (dpf) and compared standard feed conditions (StF) to a high fat diet (HFD) or high glucose diet (HGD). We also exposed zebrafish embryos to a non-toxic concentration of tributyltin (TBT, 1 nM) or Tris(1,3-dichloroisopropyl)phosphate (TDCiPP, 0.5 µM) from 0-6 dpf and reared larvae to 15 dpf under StF. Potential molecular mechanisms of altered adipogenesis were examined by qPCR. Diet-dependent modulation of adipogenesis was observed, with HFD resulting in a threefold increase in larvae with adipocytes, compared to StF and HGD. Developmental exposure to TBT but not TDCiPP significantly increased adipocyte differentiation. The expression of adipogenic genes such as pparda, lxr and lepa was altered in response to HFD or chemicals. This study shows that SRS microscopy can be successfully applied to zebrafish to visualize and quantify adipogenesis, and is a powerful approach for identifying obesogenic chemicals in vivo.
Collapse
Affiliation(s)
- Marjo J den Broeder
- Institute of Environmental, Health and Societies, Brunel University, UB8 3PH London, UK.
- Institute for Environmental Studies, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Miriam J B Moester
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Jorke H Kamstra
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. BOX 8146, Dep 0033 Oslo, Norway.
| | - Peter H Cenijn
- Institute for Environmental Studies, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Valentina Davidoiu
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
- Rotterdam Ophthalmic Institute, Rotterdam Eye Hospital, 3011 BH Rotterdam, The Netherlands.
| | - Leonie M Kamminga
- Radboud University Nijmegen, Faculty of Science, Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands.
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands.
| | - Freek Ariese
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Johannes F de Boer
- Institute for Lasers, Life and Biophotonics, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| | - Juliette Legler
- Institute of Environmental, Health and Societies, Brunel University, UB8 3PH London, UK.
- Institute for Environmental Studies, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
138
|
Dani C, Pfeifer A. The complexity of PDGFR signaling: regulation of adipose progenitor maintenance and adipocyte-myofibroblast transition. Stem Cell Investig 2017; 4:28. [PMID: 28529943 DOI: 10.21037/sci.2017.04.02] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/10/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Christian Dani
- Université Côte d'Azur, CNRS, Inserm, iBV, Faculté de Médecine 06107 Nice Cedex 2, France
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
139
|
Maurizi G, Poloni A, Mattiucci D, Santi S, Maurizi A, Izzi V, Giuliani A, Mancini S, Zingaretti MC, Perugini J, Severi I, Falconi M, Vivarelli M, Rippo MR, Corvera S, Giordano A, Leoni P, Cinti S. Human White Adipocytes Convert Into “Rainbow” Adipocytes In Vitro. J Cell Physiol 2017; 232:2887-2899. [PMID: 27987321 DOI: 10.1002/jcp.25743] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 12/15/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Giulia Maurizi
- Dipartimento Scienze Cliniche e Molecolari; Clinica di Ematologia; Università Politecnica delle Marche; Ancona Italy
| | - Antonella Poloni
- Dipartimento Scienze Cliniche e Molecolari; Clinica di Ematologia; Università Politecnica delle Marche; Ancona Italy
| | - Domenico Mattiucci
- Dipartimento Scienze Cliniche e Molecolari; Clinica di Ematologia; Università Politecnica delle Marche; Ancona Italy
| | - Spartaco Santi
- Istituto di Genetica Molecolare del CNR; Laboratorio di Biologia Cellulare Muscoloscheletrica, Istituti Ortopedici Rizzoli; Bologna Italy
| | - Angela Maurizi
- Dipartimento di Medicina Sperimentale e Clinica; Clinica Chirurgia del Pancreas; Università Politecnica delle Marche; Ancona Italy
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine; Center for Cell-Matrix Research and Biocenter Oulu; University of Oulu; Oulu Finland
| | - Angelica Giuliani
- Dipartimento Scienze Cliniche e Molecolari; Laboratorio di Patologia Sperimentale; Ancona Italy
| | - Stefania Mancini
- Dipartimento Scienze Cliniche e Molecolari; Clinica di Ematologia; Università Politecnica delle Marche; Ancona Italy
| | - Maria Cristina Zingaretti
- Dipartimento di Medicina Sperimentale e Clinica; Center of Obesity; Università Politecnica delle Marche; Ancona Italy
| | - Jessica Perugini
- Dipartimento di Medicina Sperimentale e Clinica; Center of Obesity; Università Politecnica delle Marche; Ancona Italy
| | - Ilenia Severi
- Dipartimento di Medicina Sperimentale e Clinica; Center of Obesity; Università Politecnica delle Marche; Ancona Italy
| | - Massimo Falconi
- Dipartimento di Medicina Sperimentale e Clinica; Clinica Chirurgia del Pancreas; Università Politecnica delle Marche; Ancona Italy
| | - Marco Vivarelli
- Department of Experimental and Clinical Medicine; Hepatobiliary and Abdominal Transplantation Surgery; Università Politecnica delle Marche; Ancona Italy
| | - Maria Rita Rippo
- Dipartimento Scienze Cliniche e Molecolari; Laboratorio di Patologia Sperimentale; Ancona Italy
| | - Silvia Corvera
- Program in Molecular Medicine; University of Massachusetts Medical School; Worcester Massachusetts
| | - Antonio Giordano
- Dipartimento di Medicina Sperimentale e Clinica; Center of Obesity; Università Politecnica delle Marche; Ancona Italy
| | - Pietro Leoni
- Dipartimento Scienze Cliniche e Molecolari; Clinica di Ematologia; Università Politecnica delle Marche; Ancona Italy
| | - Saverio Cinti
- Dipartimento di Medicina Sperimentale e Clinica; Center of Obesity; Università Politecnica delle Marche; Ancona Italy
| |
Collapse
|
140
|
Liu J, Xu Z, Wu W, Wang Y, Shan T. CreRecombinase Strains Used for the Study of Adipose Tissues and Adipocyte Progenitors. J Cell Physiol 2017; 232:2698-2703. [DOI: 10.1002/jcp.25675] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/01/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Jiaqi Liu
- College of Animal Sciences; Zhejiang University; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education; Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang China
| | - Ziye Xu
- College of Animal Sciences; Zhejiang University; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education; Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang China
| | - Weiche Wu
- College of Animal Sciences; Zhejiang University; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education; Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang China
| | - Yizhen Wang
- College of Animal Sciences; Zhejiang University; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education; Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang China
| | - Tizhong Shan
- College of Animal Sciences; Zhejiang University; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education; Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang China
| |
Collapse
|
141
|
Kiehn JT, Tsang AH, Heyde I, Leinweber B, Kolbe I, Leliavski A, Oster H. Circadian Rhythms in Adipose Tissue Physiology. Compr Physiol 2017; 7:383-427. [PMID: 28333377 DOI: 10.1002/cphy.c160017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The different types of adipose tissues fulfill a wide range of biological functions-from energy storage to hormone secretion and thermogenesis-many of which show pronounced variations over the course of the day. Such 24-h rhythms in physiology and behavior are coordinated by endogenous circadian clocks found in all tissues and cells, including adipocytes. At the molecular level, these clocks are based on interlocked transcriptional-translational feedback loops comprised of a set of clock genes/proteins. Tissue-specific clock-controlled transcriptional programs translate time-of-day information into physiologically relevant signals. In adipose tissues, clock gene control has been documented for adipocyte proliferation and differentiation, lipid metabolism as well as endocrine function and other adipose oscillations are under control of systemic signals tied to endocrine, neuronal, or behavioral rhythms. Circadian rhythm disruption, for example, by night shift work or through genetic alterations, is associated with changes in adipocyte metabolism and hormone secretion. At the same time, adipose metabolic state feeds back to central and peripheral clocks, adjusting behavioral and physiological rhythms. In this overview article, we summarize our current knowledge about the crosstalk between circadian clocks and energy metabolism with a focus on adipose physiology. © 2017 American Physiological Society. Compr Physiol 7:383-427, 2017.
Collapse
Affiliation(s)
- Jana-Thabea Kiehn
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Anthony H Tsang
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Isabel Heyde
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Brinja Leinweber
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Isa Kolbe
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| | - Alexei Leliavski
- Institute of Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Henrik Oster
- Chronophysiology Group, Medical Department I, University of Lübeck, Lübeck, Germany
| |
Collapse
|
142
|
Graus-Nunes F, Rachid TL, de Oliveira Santos F, Barbosa-da-Silva S, Souza-Mello V. AT1 receptor antagonist induces thermogenic beige adipocytes in the inguinal white adipose tissue of obese mice. Endocrine 2017; 55:786-798. [PMID: 28012150 DOI: 10.1007/s12020-016-1213-1] [Citation(s) in RCA: 18] [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: 09/02/2016] [Accepted: 12/15/2016] [Indexed: 01/06/2023]
Abstract
PURPOSE To evaluate whether losartan is able to induce beige adipocytes formation, focusing on the thermogenic gene expression and adipocyte remodeling in the subcutaneous white adipose tissue of diet-induced obese mice. METHODS Male C57BL/6 mice received a control diet (10% energy as lipids) or a high-fat diet (50% energy as lipids) for 10 weeks, followed by a 5-week treatment with losartan: control group, control-losartan group (10 mg/Kg/day), high-fat group and high-fat-losartan group (10 mg/Kg/day). Biochemical, morphometrical, stereological and molecular approaches were used to evaluate the outcomes. RESULTS The high-fat diet elicited overweight, insulin resistance and adipocyte hypertrophy in the high-fat group, all of which losartan rescued in the high-fat-losartan group. These effects comply with the induction of beige adipocytes within the inguinal fat pads in high-fat-losartan group as they exhibited the greatest energy expenditure among the groups along with the presence uncoupling protein 1 positive multilocular adipocytes with enhanced peroxisome proliferator-activated receptor gamma coactivator 1-alpha and PR domain containing 16 mRNA levels, indicating a significant potential for mitochondrial biogenesis and adaptive thermogenesis. CONCLUSIONS Our results show compelling evidence that losartan countered diet-induced obesity in mice by enhancing energy expenditure through beige adipocytes induction. Reduced body mass, increased insulin sensitivity, decreased adipocyte size and marked expression of uncoupling protein 1 by ectopic multilocular adipocytes support these findings. The use of losartan as a coadjutant medicine to tackle obesity and its related disorders merits further investigation.
Collapse
Affiliation(s)
- Francielle Graus-Nunes
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tamiris Lima Rachid
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe de Oliveira Santos
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sandra Barbosa-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
143
|
Porras DP, Abbaszadeh M, Bhattacharya D, D'Souza NC, Edjiu NR, Perry CGR, Scimè A. p107 Determines a Metabolic Checkpoint Required for Adipocyte Lineage Fates. Stem Cells 2017; 35:1378-1391. [PMID: 28233396 DOI: 10.1002/stem.2576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/07/2017] [Indexed: 12/14/2022]
Abstract
We show that the transcriptional corepressor p107 orchestrates a metabolic checkpoint that determines adipocyte lineage fates for non-committed progenitors. p107 accomplishes this when stem cell commitment would normally occur in growth arrested cells. p107-deficient embryonic progenitors are characterized by a metabolic state resembling aerobic glycolysis that is necessary for their pro-thermogenic fate. Indeed, during growth arrest they have a reduced capacity for NADH partitioning between the cytoplasm and mitochondria. Intriguingly, this occurred despite an increase in the capacity for mitochondrial oxidation of non-glucose substrates. The significance of metabolic reprogramming is underscored by the disruption of glycolytic capacities in p107-depleted progenitors that reverted their fates from pro-thermogenic to white adipocytes. Moreover, the manipulation of glycolytic capacity on nonspecified embryonic and adult progenitors forced their beige fat commitment. These innovative findings introduce a new approach to increase pro-thermogenic adipocytes based on simply promoting aerobic glycolysis to manipulate nonspecified progenitor fate decisions. Stem Cells 2017;35:1378-1391.
Collapse
Affiliation(s)
- Deanna P Porras
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Maryam Abbaszadeh
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Debasmita Bhattacharya
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Ninoschka C D'Souza
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Nareh R Edjiu
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Christopher G R Perry
- Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| | - Anthony Scimè
- Stem Cell Research Group, York University, Toronto, Ontario, Canada.,Molecular, Cellular and Integrative Physiology, Faculty of Health, York University, Toronto, Ontario, Canada
| |
Collapse
|
144
|
Invited review: Pre- and postnatal adipose tissue development in farm animals: from stem cells to adipocyte physiology. Animal 2017; 10:1839-1847. [PMID: 27751202 DOI: 10.1017/s1751731116000872] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Both white and brown adipose tissues are recognized to be differently involved in energy metabolism and are also able to secrete a variety of factors called adipokines that are involved in a wide range of physiological and metabolic functions. Brown adipose tissue is predominant around birth, except in pigs. Irrespective of species, white adipose tissue has a large capacity to expand postnatally and is able to adapt to a variety of factors. The aim of this review is to update the cellular and molecular mechanisms associated with pre- and postnatal adipose tissue development with a special focus on pigs and ruminants. In contrast to other tissues, the embryonic origin of adipose cells remains the subject of debate. Adipose cells arise from the recruitment of specific multipotent stem cells/progenitors named adipose tissue-derived stromal cells. Recent studies have highlighted the existence of a variety of those cells being able to differentiate into white, brown or brown-like/beige adipocytes. After commitment to the adipocyte lineage, progenitors undergo large changes in the expression of many genes involved in cell cycle arrest, lipid accumulation and secretory functions. Early nutrition can affect these processes during fetal and perinatal periods and can also influence or pre-determinate later growth of adipose tissue. How these changes may be related to adipose tissue functional maturity around birth and can influence newborn survival is discussed. Altogether, a better knowledge of fetal and postnatal adipose tissue development is important for various aspects of animal production, including neonatal survival, postnatal growth efficiency and health.
Collapse
|
145
|
Aldiss P, Dellschaft N, Sacks H, Budge H, Symonds ME. Beyond obesity – thermogenic adipocytes and cardiometabolic health. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0007/hmbci-2017-0007.xml. [DOI: 10.1515/hmbci-2017-0007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 03/22/2017] [Indexed: 12/22/2022]
Abstract
AbstractThe global prevalence of obesity and related cardiometabolic disease continues to increase through the 21st century. Whilst multi-factorial, obesity is ultimately caused by chronic caloric excess. However, despite numerous interventions focussing on reducing caloric intake these either fail or only elicit short-term changes in body mass. There is now a focus on increasing energy expenditure instead which has stemmed from the recent ‘re-discovery’ of cold-activated brown adipose tissue (BAT) in adult humans and inducible ‘beige’ adipocytes. Through the unique mitochondrial uncoupling protein 1 (UCP1), these thermogenic adipocytes are capable of combusting large amounts of chemical energy as heat and in animal models can prevent obesity and cardiometabolic disease. At present, human data does not point to a role for thermogenic adipocytes in regulating body weight or fat mass but points to a pivotal role in regulating metabolic health by improving insulin resistance as well as glucose and lipid homeostasis. This review will therefore focus on the metabolic benefits of BAT activation and the mechanisms and signalling pathways by which these could occur including improvements in insulin signalling in peripheral tissues, systemic lipid and cholesterol metabolism and cardiac and vascular function.
Collapse
|
146
|
Chen M, Liang S, Zhou H, Xu Y, Qin X, Hu Z, Wang X, Qiu L, Wang W, Zhang Y, Ying Z. Prenatal and postnatal mothering by diesel exhaust PM 2.5-exposed dams differentially program mouse energy metabolism. Part Fibre Toxicol 2017; 14:3. [PMID: 28100227 PMCID: PMC5423412 DOI: 10.1186/s12989-017-0183-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/11/2017] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Obesity is one of the leading threats to global public health. It is consequent to abnormal energy metabolism. Currently, it has been well established that maternal exposure to environmental stressors that cause inappropriate fetal development may have long-term adverse effects on offspring energy metabolism in an exposure timing-dependent manner, known as developmental programming of health and diseases paradigm. Rapidly increasing evidence has indicated that maternal exposure to ambient fine particles (PM2.5) correlates to abnormal fetal development. In the present study, we therefore assessed whether maternal exposure to diesel exhaust PM2.5 (DEP), the major component of ambient PM2.5 in urban areas, programs offspring energy metabolism, and further examined how the timing of exposure impacts this programming. RESULTS The growth trajectory of offspring shows that although prenatal maternal exposure to DEP did not impact the birth weight of offspring, it significantly decreased offspring body weight from postnatal week 2 until the end of observation. This weight loss effect of prenatal maternal exposure to DEP coincided with decreased food intake but not alteration in brown adipose tissue (BAT) morphology. The hypophagic effect of prenatal maternal exposure to DEP was in concord with decreased hypothalamic expression of an orexigenic peptide NPY, suggesting that the prenatal maternal exposure to DEP impacts offspring energy balance primarily through programming of food intake. Paradoxically, the reduced body weight resulted from prenatal maternal exposure to DEP was accompanied by increased mass of epididymal adipose tissue, which was due to hyperplasia as morphological analysis did not observe any hypertrophy. In direct contrast, the postnatal mothering by DEP-exposed dams increased offspring body weight during lactation and adulthood, paralleled by markedly increased fat accumulation and decreased UCP1 expression in BAT but not alteration in food intake. The weight gain induced by postnatal mothering by DEP-exposed dams was also expressed as an increased adiposity. But it concurred with a marked hypertrophy of adipocytes. CONCLUSION Prenatal and postnatal mothering by DEP-exposed dams differentially program offspring energy metabolism, underscoring consideration of the exposure timing when examining the adverse effects of maternal exposure to ambient PM2.5.
Collapse
Affiliation(s)
- Minjie Chen
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032 China
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
| | - Shuai Liang
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
- Department of Bile Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008 China
| | - Huifen Zhou
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
- Department of Pathology, Hubei University of Science and Technology, Xianning, Hubei 437100 China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032 China
| | - Xiaobo Qin
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
| | - Ziying Hu
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
- Department of Endocrinology, the People’s Hospital of Zhengzhou University (Henan Provincial People’s Hospital), Zhengzhou, Henan 450003 China
| | - Xiaoke Wang
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
- Department of Occupational and Environmental Health, School of Public Health, Nantong University, Nantong, 226019 China
| | - Lianglin Qiu
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
- Department of Occupational and Environmental Health, School of Public Health, Nantong University, Nantong, 226019 China
| | - Wanjun Wang
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032 China
| | - Yuhao Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032 China
| | - Zhekang Ying
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032 China
- Department of Medicine Cardiology Division, School of Medicine, University of Maryland, 20 Penn St. HSFII S022, Baltimore, MD 21201 USA
| |
Collapse
|
147
|
Bargut TCL, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Browning of white adipose tissue: lessons from experimental models. Horm Mol Biol Clin Investig 2017; 31:hmbci-2016-0051. [PMID: 28099124 DOI: 10.1515/hmbci-2016-0051] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/01/2016] [Indexed: 04/25/2024]
Abstract
Beige or brite (brown-in-white) adipocytes are present in white adipose tissue (WAT) and have a white fat-like phenotype that when stimulated acquires a brown fat-like phenotype, leading to increased thermogenesis. This phenomenon is known as browning and is more likely to occur in subcutaneous fat depots. Browning involves the expression of many transcription factors, such as PR domain containing 16 (PRDM16) and peroxisome proliferator-activated receptor (PPAR)-γ, and of uncoupling protein (UCP)-1, which is the hallmark of thermogenesis. Recent papers pointed that browning can occur in the WAT of humans, with beneficial metabolic effects. This fact indicates that these cells can be targeted to treat a range of diseases, with both pharmacological and nutritional activators. Pharmacological approaches to induce browning include the use of PPAR-α agonist, adrenergic receptor stimulation, thyroid hormone administration, irisin and FGF21 induction. Most of them act through the induction of PPAR-γ coactivator (PGC) 1-α and the consequent mitochondrial biogenesis and UCP1 induction. About the nutritional inducers, several compounds have been described with multiple mechanisms of action. Some of these activators include specific amino acids restriction, capsaicin, bile acids, Resveratrol, and retinoic acid. Besides that, some classes of lipids, as well as many plant extracts, have also been implicated in the browning of WAT. In conclusion, the discovery of browning in human WAT opens the possibility to target the adipose tissue to fight a range of diseases. Studies have arisen showing promising results and bringing new opportunities in thermogenesis and obesity control.
Collapse
Affiliation(s)
- Thereza Cristina Lonzetti Bargut
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratorio de Morfometria, Metabolismo e Doença Cardiovascular, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, 20551-030 Rio de Janeiro, Brazil, Phone (+55.21) 2868-8316, Fax: 2868-8033, E-mail:
| |
Collapse
|
148
|
Tubulin alpha 8 is expressed in hepatic stellate cells and is induced in transformed hepatocytes. Mol Cell Biochem 2017; 428:161-170. [DOI: 10.1007/s11010-016-2926-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/21/2016] [Indexed: 12/16/2022]
|
149
|
Yao F, Zhang M, Chen L. Adipose Tissue-Specialized Immunologic Features Might Be the Potential Therapeutic Target of Prospective Medicines for Obesity. J Diabetes Res 2017; 2017:4504612. [PMID: 28466023 PMCID: PMC5390594 DOI: 10.1155/2017/4504612] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/03/2017] [Accepted: 03/13/2017] [Indexed: 12/19/2022] Open
Abstract
Excessive lipid accumulation in adipose tissue is either the source of obesity or the cause and result of chronic local inflammation, and recent studies indicate that the accumulation may induce many other specialized immunologic features with macrophages and epidemic diseases. We analyze the effective stages of immune cells in adipose tissue, including macrophage recruitment, macrophage polarization, and macrophage-like phenotype preadipocyte possession to find optimal sites as drug targets. Subsequently, some main signaling pathways are summarized in this review, including the AMP-activated protein kinase (AMPK) pathway, the JNK signaling pathway, and a novel one, the Notch signaling pathway. We illustrate all these points in order to determine the general pathogenesis of chronic low-grade local inflammation in adipose tissue and the related signaling pathways. In addition, signal-associated prospective compounds, such as berberine, are summarized and discussed with potential targets in pathogenesis. This might provide some possible thoughts and novel therapies for studying chronic inflammatory diseases, such as insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Fan Yao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- School of Nursing, Jilin University, Changchun 130021, China
- *Li Chen:
| |
Collapse
|
150
|
Wu M, Liu D, Zeng R, Xian T, Lu Y, Zeng G, Sun Z, Huang B, Huang Q. Epigallocatechin-3-gallate inhibits adipogenesis through down-regulation of PPARγ and FAS expression mediated by PI3K-AKT signaling in 3T3-L1 cells. Eur J Pharmacol 2016; 795:134-142. [PMID: 27940057 DOI: 10.1016/j.ejphar.2016.12.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 01/09/2023]
Abstract
Epigallocatechin-3-gallate (EGCG), a major component in green tea, functions as extensive bioactivities including anti-inflammation, anti-oxidation, and anti-cancer. However, little is known about its anti-adipogenesis and underlying mechanisms. The purport of this study sought to investigate effects of EGCG on 3T3-L1 preadipocyte differentiation and to explore its possible mechanisms. The 3T3-L1 cells were induced to differentiate under the condition of pro-adipogenic cocktail with or without indicated EGCG concentrations (10, 50, 100, 200µM) for 2, 4, 6 and 8 days, respectively. Also, another batch of 3T3-L1 cells was induced under the optimal EGCG concentration (100µM) with or without SC3036 (PI3K activator, 10µM) or SC79 (AKT activator, 0.5µM) for 8 days. Subsequently, the cell viability was examined by MTT assay and the cell morphology was visualized by Oil red O staining. Finally, the mRNA levels including peroxisome proliferator activated receptor γ (PPARγ) and fatty acid synthase (FAS) were detected by quantitative real time PCR, while the protein levels of PPARγ, FAS, phosphatidylinositol 3 kinase (PI3K), insulin receptor substrate1(IRS1), AKT, and p-AKT were measured by immunoblotting analysis. Our results showed that EGCG inhibited adipogenesis of 3T3-L1 preadipocyte in a concentration-dependent manner. Moreover, the inhibitory effects were reversed by SC3036 or SC79, suggesting that the inhibitory effects of EGCG are mediated by PI3K-AKT signaling to down-regulate PPARγ and FAS expression levels. The findings shed light on EGCG anti-adipogenic effects and its underlying mechanism and provide a novel preventive-therapeutic potential for obesity subjects as a compound from Chinese green tea.
Collapse
Affiliation(s)
- Mengqing Wu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Dan Liu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Rong Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Tao Xian
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Yi Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Zhangzetian Sun
- Jiangxi Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Bowei Huang
- Jiangxi Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China.
| |
Collapse
|