51
|
Jiao F, Yan X, Yu Y, Zhu X, Ma Y, Yue Z, Ou H, Yan Z. Protective effects of maternal methyl donor supplementation on adult offspring of high fat diet-fed dams. J Nutr Biochem 2016; 34:42-51. [DOI: 10.1016/j.jnutbio.2016.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 01/05/2023]
|
52
|
Sinclair KD, Rutherford KMD, Wallace JM, Brameld JM, Stöger R, Alberio R, Sweetman D, Gardner DS, Perry VEA, Adam CL, Ashworth CJ, Robinson JE, Dwyer CM. Epigenetics and developmental programming of welfare and production traits in farm animals. Reprod Fertil Dev 2016; 28:RD16102. [PMID: 27439952 DOI: 10.1071/rd16102] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/06/2016] [Indexed: 12/11/2022] Open
Abstract
The concept that postnatal health and development can be influenced by events that occur in utero originated from epidemiological studies in humans supported by numerous mechanistic (including epigenetic) studies in a variety of model species. Referred to as the 'developmental origins of health and disease' or 'DOHaD' hypothesis, the primary focus of large-animal studies until quite recently had been biomedical. Attention has since turned towards traits of commercial importance in farm animals. Herein we review the evidence that prenatal risk factors, including suboptimal parental nutrition, gestational stress, exposure to environmental chemicals and advanced breeding technologies, can determine traits such as postnatal growth, feed efficiency, milk yield, carcass composition, animal welfare and reproductive potential. We consider the role of epigenetic and cytoplasmic mechanisms of inheritance, and discuss implications for livestock production and future research endeavours. We conclude that although the concept is proven for several traits, issues relating to effect size, and hence commercial importance, remain. Studies have also invariably been conducted under controlled experimental conditions, frequently assessing single risk factors, thereby limiting their translational value for livestock production. We propose concerted international research efforts that consider multiple, concurrent stressors to better represent effects of contemporary animal production systems.
Collapse
|
53
|
Isolation and Fluorescence-Activated Cell Sorting of Murine WT1-Expressing Adipocyte Precursor Cells. Methods Mol Biol 2016. [PMID: 27417961 DOI: 10.1007/978-1-4939-4023-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The current global obesity epidemic has triggered increased interest in adipose tissue biology. A major area of attention for many is adipose tissue development. A greater understanding of adipocyte ontogeny could be highly beneficial in answering questions about obesity-associated disease. Recent work has shown that a proportion of mature adipocytes in visceral white adipose tissue are derived from Wt1-expressing adipocyte precursor cells. These adipocyte precursor cells reside within the adipose tissue itself, and are a constituent of the stromal vascular fraction (SVF), along with other, non-adipogenic, cell types. Crucially, heterogeneity exists within the adipocyte precursor population, with only a proportion of cells expressing Wt1. Moreover, it appears that this difference in the precursor cells may influence the mature adipocytes, with Wt1-lineage-positive adipocytes having fewer, larger lipid droplets than the Wt1-lineage negative. Using fluorescence-activated cell sorting, based on specific marker profiles, it is possible to isolate the adipocyte precursor cells from the SVF. Subsequently, this population can be divided into Wt1-expressing and non-expressing fractions, therefore permitting further analysis of the two cell populations, and the mature adipocytes derived from them. In this chapter we outline a method by which adipocyte precursor cells can be isolated, and how, using a specific mouse model, Wt1-expressing and non-expressing cells can be separated.
Collapse
|
54
|
Gimble JM, Ray SP, Zanata F, Wu X, Wade J, Khoobehi K, Ferreira LM, Bunnell BA. Adipose Derived Cells and Tissues for Regenerative Medicine. ACS Biomater Sci Eng 2016; 3:1477-1482. [DOI: 10.1021/acsbiomaterials.6b00261] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
| | - Stephen P. Ray
- Cancer
Treatment Center, Midwestern Regional Medical Center, Zion, Illinois 60099, United States
| | - Fabiana Zanata
- Universidade Federal São Paulo, São
Paulo 04021-001, Brazil
| | - Xiying Wu
- LaCell LLC, New Orleans, Louisiana 70112, United States
| | - James Wade
- Plastic Surgery Consultants, Baton Rouge, Louisiana 70808, United States
| | - Kamran Khoobehi
- Khoobehi and Associates, Metairie, Louisiana 70002, United States
| | | | - Bruce A. Bunnell
- Department
of Pharmacology, Tulane University School of Medicine, New Orleans Louisiana 70112, United States
- Tulane National Primate Research Center, Covington, Louisiana 70433, United States
| |
Collapse
|
55
|
Microenvironmental Control of Adipocyte Fate and Function. Trends Cell Biol 2016; 26:745-755. [PMID: 27268909 DOI: 10.1016/j.tcb.2016.05.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/09/2016] [Accepted: 05/16/2016] [Indexed: 01/07/2023]
Abstract
The properties of tissue-specific microenvironments vary widely in the human body and demonstrably influence the structure and function of many cell types. Adipocytes are no exception, responding to cues in specialized niches to perform vital metabolic and endocrine functions. The adipose microenvironment is remodeled during tissue expansion to maintain the structural and functional integrity of the tissue and disrupted remodeling in obesity contributes to the progression of metabolic syndrome, breast cancer, and other malignancies. The increasing incidence of these obesity-related diseases and the recent focus on improved in vitro models of human tissue biology underscore growing interest in the regulatory role of adipocyte microenvironments in health and disease.
Collapse
|
56
|
Salazar J, Luzardo E, Mejías JC, Rojas J, Ferreira A, Rivas-Ríos JR, Bermúdez V. Epicardial Fat: Physiological, Pathological, and Therapeutic Implications. Cardiol Res Pract 2016; 2016:1291537. [PMID: 27213076 PMCID: PMC4861775 DOI: 10.1155/2016/1291537] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 03/02/2016] [Accepted: 04/03/2016] [Indexed: 12/20/2022] Open
Abstract
Epicardial fat is closely related to blood supply vessels, both anatomically and functionally, which is why any change in this adipose tissue's behavior is considered a potential risk factor for cardiovascular disease development. When proinflammatory adipokines are released from the epicardial fat, this can lead to a decrease in insulin sensitivity, low adiponectin production, and an increased proliferation of vascular smooth muscle cells. These adipokines move from one compartment to another by either transcellular passing or diffusion, thus having the ability to regulate cardiac muscle activity, a phenomenon called vasocrine regulation. The participation of these adipokines generates a state of persistent vasoconstriction, increased stiffness, and weakening of the coronary wall, consequently contributing to the formation of atherosclerotic plaques. Therefore, epicardial adipose tissue thickening should be considered a risk factor in the development of cardiovascular disease, a potential therapeutic target for cardiovascular pathology and a molecular point of contact for "endocrine-cardiology."
Collapse
Affiliation(s)
- Juan Salazar
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - Eliana Luzardo
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - José Carlos Mejías
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - Joselyn Rojas
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Antonio Ferreira
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
- Internal Medicine Service, “Dr. Manuel Noriega Trigo” Hospital, San Francisco 4004, Venezuela
| | - José Ramón Rivas-Ríos
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| | - Valmore Bermúdez
- Endocrine and Metabolic Diseases Research Center, University of Zulia, Maracaibo 4004, Venezuela
| |
Collapse
|
57
|
Berryman DE, Henry B, Hjortebjerg R, List EO, Kopchick JJ. Developments in our understanding of the effects of growth hormone on white adipose tissue from mice: implications to the clinic. Expert Rev Endocrinol Metab 2016; 11:197-207. [PMID: 28435436 PMCID: PMC5397118 DOI: 10.1586/17446651.2016.1147950] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adipose tissue (AT) is a well-established target of growth hormone (GH) and is altered in clinical conditions associated with excess, deficiency and absence of GH action. Due to the difficulty in collecting AT from clinical populations, genetically modified mice have been useful in better understanding how GH affects this tissue. Recent findings in mice would suggest that the impact of GH on AT is beyond alterations of lipolysis, lipogenesis or proliferation/ differentiation. AT depot-specific alterations in immune cells, extracellular matrix, adipokines, and senescence indicate an expanded role for GH in AT physiology. This mouse data will guide additional studies necessary to evaluate the therapeutic potential and safety of GH for conditions associated with altering AT, such as obesity. In this review, we introduce several relatively new intricacies of GH's effect on AT, focusing on recent studies in mice. Finally, we summarize the clinical implications of these findings.
Collapse
Affiliation(s)
- Darlene E Berryman
- Executive Director, The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, (740) 593-9661 - phone, (740) 593-4795 - fax
| | - Brooke Henry
- 108 Konneker Research Labs, Ohio University, (740) 593-9665
| | - Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University, Noerrebrogade 44, 8000 Aarhus C, Denmark, +45 6166 8045 - phone, +45 7846 2150 - fax
| | - Edward O List
- Senior Scientist, 218 Konneker Research Labs, Edison Biotechnology Institute, Ohio University, (740) 593-4620 - phone, (740) 593-4795 - fax
| | - John J Kopchick
- Distinguished Professor, Goll Ohio Eminent Scholar, 172 Water Tower Drive, Ohio University, (740) 593-4534 - phone, (740) 593-4795 - fax
| |
Collapse
|
58
|
Durandt C, van Vollenstee FA, Dessels C, Kallmeyer K, de Villiers D, Murdoch C, Potgieter M, Pepper MS. Novel flow cytometric approach for the detection of adipocyte subpopulations during adipogenesis. J Lipid Res 2016; 57:729-42. [PMID: 26830859 PMCID: PMC4808761 DOI: 10.1194/jlr.d065664] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Indexed: 12/17/2022] Open
Abstract
The ability of mesenchymal stromal cells (MSCs) to differentiate into adipocytes provides a cellular model of human origin to study adipogenesis in vitro. One of the major challenges in studying adipogenesis is the lack of tools to identify and monitor the differentiation of various subpopulations within the heterogeneous pool of MSCs. Cluster of differentiation (CD)36 plays an important role in the formation of intracellular lipid droplets, a key characteristic of adipocyte differentiation/maturation. The objective of this study was to develop a reproducible quantitative method to study adipocyte differentiation by comparing two lipophilic dyes [Nile Red (NR) and Bodipy 493/503] in combination with CD36 surface marker staining. We identified a subpopulation of adipose-derived stromal cells that express CD36 at intermediate/high levels and show that combining CD36 cell surface staining with neutral lipid-specific staining allows us to monitor differentiation of adipose-derived stromal cells that express CD36intermediate/high during adipocyte differentiation in vitro. The gradual increase of CD36intermediate/high/NRpositive cells during the 21 day adipogenesis induction period correlated with upregulation of adipogenesis-associated gene expression.
Collapse
Affiliation(s)
- Chrisna Durandt
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Fiona A van Vollenstee
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Carla Dessels
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Karlien Kallmeyer
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Danielle de Villiers
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Candice Murdoch
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Marnie Potgieter
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S Pepper
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
59
|
Li C, Zhou L. Inhibitory effect 6-gingerol on adipogenesis through activation of the Wnt/β-catenin signaling pathway in 3T3-L1 adipocytes. Toxicol In Vitro 2015; 30:394-401. [PMID: 26498061 DOI: 10.1016/j.tiv.2015.09.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 09/19/2015] [Accepted: 09/24/2015] [Indexed: 11/26/2022]
Abstract
6-Gingerol has been reported to inhibit adipogenesis and lipid content accumulation. However, the mechanism of its anti-adipogenic effect remains unclear. Our aim is to investigate the molecular mechanism of the anti-adipogenic effect of 6-gingerol. The lipid content in adipocytes was measured by Oil Red O staining and cell viability was analyzed by MTT assay. The extent of suppression of differentiation by 6-gingerol was characterized by measuring the triglyceride content and GPDH activity. The regulation of adipogenic markers and the components of the Wnt/β-catenin pathway were analyzed by real-time PCR and Western blotting. The nuclear location of β-catenin was identified using immunofluorescence assay. Small interfering RNA transfection was conducted to elucidate the crucial role of β-catenin in anti-adipogenic effect of 6-gingerol. Our results showed that 6-gingerol inhibited the adipogenesis and lowered the mRNA expression levels of transcription factors and the key lipogenic enzymes in 3T3-L1 cells. The effect of 6-gingerol on adipogenic differentiation was accompanied by stimulating the activation of the Wnt/β-catenin signaling. In addition, we found that 6-gingerol induced phosphorylations of glycogen synthase kinase-3β(GSK-3β), and promoted the nuclear accumulation of β-catenin. Importantly, the inhibitory effect of 6-gingerol on adipogenic differentiation was reversed after the siRNA knockdown of β-catenin was added. Our findings demonstrated that 6-gingerol inhibits the adipogenic differentiation of 3T3-L1 cells through activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Chunbo Li
- Department of Gynaecology and Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China.
| | - Lin Zhou
- Department of urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
60
|
Gupta OT, Gupta RK. Visceral Adipose Tissue Mesothelial Cells: Living on the Edge or Just Taking Up Space? Trends Endocrinol Metab 2015; 26:515-523. [PMID: 26412153 DOI: 10.1016/j.tem.2015.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/29/2015] [Accepted: 07/13/2015] [Indexed: 01/22/2023]
Abstract
Visceral adiposity and pathological adipose tissue remodeling, a result of overnutrition, are strong predictors of metabolic health in obesity. Factors intrinsic to visceral adipose depots are likely to play a causal role in eliciting the detrimental effects of this tissue on systemic nutrient homeostasis. The visceral adipose-associated mesothelium, a monolayer of epithelial cells of mesodermal origin that line the visceral serosa, has recently attracted attention for its role in metabolic dysfunction. Here we highlight and consolidate literature from various fields of study that points to the visceral adipose-associated mesothelium as a potential contributor to adipose development and remodeling. We propose a hypothesis in which adipose mesothelial cells represent a visceral depot-specific determinant of adipose tissue health in obesity.
Collapse
Affiliation(s)
- Olga T Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, Division of Pediatric Endocrinology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
61
|
Chau YY, Hastie N. Wt1, the mesothelium and the origins and heterogeneity of visceral fat progenitors. Adipocyte 2015; 4:217-21. [PMID: 26257994 PMCID: PMC4496970 DOI: 10.4161/21623945.2014.985009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 01/22/2023] Open
Abstract
One major gap in adipocyte biology has been a lack of understanding of the developmental origins of the different visceral white adipose tissue (WAT) depots and subcutaneous WAT. In a recent study we showed that most visceral WAT but no subcutaneous WAT arises from cells expressing the Wilms’ tumor 1 (Wt1) gene late in mouse gestation.1 Wt1 continues to be expressed in visceral WAT progenitors into adult life. We also showed that visceral WAT is lined by a mesothelium and provided evidence that this structure is the source of adipocytes. Our study also adds to the growing body of evidence that there is heterogeneity in the visceral progenitors, such that there are Wt1-expressing and non-expressing subsets, the relative proportions of which vary between depots. This raises the enticing prospect that the adipocytes arising from these progenitor subsets may have different properties and our preliminary data support this notion. Finally, evidence from our study and one from Spiegelman's group2 suggests that Wt1 is not just a marker but regulates visceral WAT identity and the progenitor population. We discuss the implications of this work and some of the questions and future directions that arise from it.
Collapse
|
62
|
Stefanon B, Pomari E, Colitti M. Effects of Rosmarinus officinalis extract on human primary omental preadipocytes and adipocytes. Exp Biol Med (Maywood) 2015; 240:884-95. [PMID: 25710930 DOI: 10.1177/1535370214562341] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/12/2014] [Indexed: 12/13/2022] Open
Abstract
The prevalence of obesity is increasing all over the world. Although it has been shown that natural substances influence fat metabolism, little is known about the effect on cellular and molecular mechanisms in human. In this in vitro study, the activity of Rosmarinus officinalis (RO) standardized extract in modulating human primary visceral preadipocytes differentiation, lipolysis, and apoptosis was investigated. Moreover, gene expression of key adipogenesis modulators and microRNAs-seq were evaluated. Preadipocytes treated with RO extract significantly reduced triglyceride incorporation during maturation in a dose-dependent manner without affecting cell viability. In addition, RO extract stimulated lipolytic activity in differentiating preadipocytes and mature adipocytes in treated cells compared to controls. Differentiating preadipocytes incubated in the presence of RO extract showed a decreased expression of cell cycle genes such as cyclin D1, cyclin-dependent kinase 4, cyclin-dependent kinase inhibitor 1A (p21, Cip1) and an increased expression of GATA binding protein 3, wingless-type MMTV integration site family, member 3A mRNA levels. Recent studies have demonstrated that some phytochemicals alter the expression of specific genes and microRNAs that play a fundamental role in the pathogenesis of obesity and related diseases. Interestingly, genes modulated in RO-treated cells were found to be validated miRNAs targets, such as let-7f-1, miR-17, and miR-143. The results indicated that RO extract modulates human adipocyte differentiation and significantly interferes with adipogenesis and lipid metabolism, supporting its interest as dietary supplement.
Collapse
Affiliation(s)
- Bruno Stefanon
- Department of Agricultural and Environmental Sciences, University of Udine, 33100 Udine, Italy
| | - Elena Pomari
- Department of Agricultural and Environmental Sciences, University of Udine, 33100 Udine, Italy
| | - Monica Colitti
- Department of Agricultural and Environmental Sciences, University of Udine, 33100 Udine, Italy
| |
Collapse
|
63
|
Hilton C, Karpe F, Pinnick KE. Role of developmental transcription factors in white, brown and beige adipose tissues. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:686-96. [PMID: 25668679 DOI: 10.1016/j.bbalip.2015.02.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/08/2015] [Accepted: 02/03/2015] [Indexed: 02/06/2023]
Abstract
In this review we discuss the role of developmental transcription factors in adipose tissue biology with a focus on how these developmental genes may contribute to regional variation in adipose tissue distribution and function. Regional, depot-specific, differences in lipid handling and signalling (lipolysis, lipid storage and adipokine/lipokine signalling) are important determinants of metabolic health. At a cellular level, preadipocytes removed from their original depot and cultured in vitro retain depot-specific functional properties, implying that these are intrinsic to the cells and not a function of their environment in situ. High throughput screening has identified a number of developmental transcription factors involved in embryological development, including members of the Homeobox and T-Box gene families, that are strongly differentially expressed between regional white adipose tissue depots and also between brown and white adipose tissue. However, the significance of depot-specific developmental signatures remains unclear. Developmental transcription factors determine body patterning during embryogenesis. The divergent developmental origins of regional adipose tissue depots may explain their differing functional characteristics. There is evidence from human genetics that developmental genes determine adipose tissue distribution: in GWAS studies a number of developmental genes have been identified as being correlated with anthropometric measures of adiposity and fat distribution. Additionally, compelling functional studies have recently implicated developmental genes in both white adipogenesis and the so-called 'browning' of white adipose tissue. Understanding the genetic and developmental pathways in adipose tissue may help uncover novel ways to intervene with the function of adipose tissue in order to promote health.
Collapse
Affiliation(s)
- Catriona Hilton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, OUH Trust, Churchill Hospital, Oxford, UK
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
64
|
Loh NY, Neville MJ, Marinou K, Hardcastle SA, Fielding BA, Duncan EL, McCarthy MI, Tobias JH, Gregson CL, Karpe F, Christodoulides C. LRP5 regulates human body fat distribution by modulating adipose progenitor biology in a dose- and depot-specific fashion. Cell Metab 2015; 21:262-273. [PMID: 25651180 PMCID: PMC4321886 DOI: 10.1016/j.cmet.2015.01.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/08/2014] [Accepted: 01/14/2015] [Indexed: 12/29/2022]
Abstract
Common variants in WNT pathway genes have been associated with bone mass and fat distribution, the latter predicting diabetes and cardiovascular disease risk. Rare mutations in the WNT co-receptors LRP5 and LRP6 are similarly associated with bone and cardiometabolic disorders. We investigated the role of LRP5 in human adipose tissue. Subjects with gain-of-function LRP5 mutations and high bone mass had enhanced lower-body fat accumulation. Reciprocally, a low bone mineral density-associated common LRP5 allele correlated with increased abdominal adiposity. Ex vivo LRP5 expression was higher in abdominal versus gluteal adipocyte progenitors. Equivalent knockdown of LRP5 in both progenitor types dose-dependently impaired β-catenin signaling and led to distinct biological outcomes: diminished gluteal and enhanced abdominal adipogenesis. These data highlight how depot differences in WNT/β-catenin pathway activity modulate human fat distribution via effects on adipocyte progenitor biology. They also identify LRP5 as a potential pharmacologic target for the treatment of cardiometabolic disorders.
Collapse
Affiliation(s)
- Nellie Y Loh
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford OX3 7LE, UK
| | - Kyriakoula Marinou
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; Department of Experimental Physiology, Athens University School of Medicine, Athens 11527, Greece
| | - Sarah A Hardcastle
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol BS10 5NB, UK
| | - Barbara A Fielding
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Emma L Duncan
- University of Queensland Diamantina Institute, School of Medicine and University of Queensland Centre for Clinical Research, Faculty of Medicine and Biomedical Sciences, University of Queensland, Woolloongabba, QLD 4102, Australia; Department of Endocrinology, Royal Brisbane and Women's Hospital, Butterfield Street, Herston, QLD 4029, Australia
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford OX3 7LE, UK
| | - Jonathan H Tobias
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol BS10 5NB, UK
| | - Celia L Gregson
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol BS10 5NB, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford OX3 7LE, UK.
| | - Constantinos Christodoulides
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK.
| |
Collapse
|
65
|
Characterization of human knee and chin adipose-derived stromal cells. Stem Cells Int 2015; 2015:592090. [PMID: 25733979 PMCID: PMC4334981 DOI: 10.1155/2015/592090] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/09/2015] [Indexed: 01/29/2023] Open
Abstract
Animal study findings have revealed that individual fat depots are not functionally equivalent and have different embryonic origins depending on the anatomic location. Mouse bone regeneration studies have also shown that it is essential to match the Hox code of transplanted cells and host tissues to achieve correct repair. However, subcutaneous fat depots from any donor site are often used in autologous fat grafting. Our study was thus carried out to determine the embryonic origins of human facial (chin) and limb (knee) fat depots and whether they had similar features and molecular matching patterns. Paired chin and knee fat depots were harvested from 11 subjects and gene expression profiles were determined by DNA microarray analyses. Adipose-derived stromal cells (ASCs) from both sites were isolated and analyzed for their capacity to proliferate, form clones, and differentiate. Chin and knee fat depots expressed a different HOX code and could have different embryonic origins. ASCs displayed a different phenotype, with chin-ASCs having the potential to differentiate into brown-like adipocytes, whereas knee-ASCs differentiated into white adipocytes. These results highlighted different features for these two fat sites and indicated that donor site selection might be an important factor to be considered when applying adipose tissue in cell-based therapies.
Collapse
|
66
|
Abstract
The distribution of adipose tissue in the body has wide-ranging and reproducible associations with health and disease. Accumulation of adipose tissue in the upper body (abdominal obesity) is associated with the development of cardiovascular disease, insulin resistance, type 2 diabetes mellitus and even all-cause mortality. Conversely, accumulation of fat in the lower body (gluteofemoral obesity) shows opposite associations with cardiovascular disease and type 2 diabetes mellitus when adjusted for overall fat mass. The abdominal depots are characterized by rapid uptake of predominantly diet-derived fat and a high lipid turnover that is easily stimulated by adrenergic receptor activation. The lower-body fat stores have a reduced lipid turnover with a capacity to accommodate fat undergoing redistribution. Lower-body adipose tissue also seems to retain the capacity to recruit additional adipocytes as a result of weight gain and demonstrates fewer signs of inflammatory insult. New data suggest that the profound functional differences between the upper-body and lower-body tissues are controlled by site-specific sets of developmental genes, such as HOXA6, HOXA5, HOXA3, IRX2 and TBX5 in subcutaneous abdominal adipose tissue and HOTAIR, SHOX2 and HOXC11 in gluteofemoral adipose tissue, which are under epigenetic control. This Review discusses the developmental and functional differences between upper-body and lower-body fat depots and provides mechanistic insight into the disease-protective effects of lower-body fat.
Collapse
Affiliation(s)
- Fredrik Karpe
- 1] Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Headington OX3 7LE, UK. [2] NIHR Oxford Biomedical Research Centre, OUH Trust, Churchill Hospital, Headington OX3 7LE, UK
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Headington OX3 7LE, UK
| |
Collapse
|
67
|
Mohsen-Kanson T, Hafner AL, Wdziekonski B, Takashima Y, Villageois P, Carrière A, Svensson M, Bagnis C, Chignon-Sicard B, Svensson PA, Casteilla L, Smith A, Dani C. Differentiation of human induced pluripotent stem cells into brown and white adipocytes: role of Pax3. Stem Cells 2015; 32:1459-67. [PMID: 24302443 DOI: 10.1002/stem.1607] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/03/2013] [Indexed: 12/11/2022]
Abstract
Identification of molecular mechanisms involved in generation of different types of adipocytes is progressing substantially in mice. However, much less is known regarding characterization of brown (BAP) and white adipocyte progenitors (WAPs) in humans, highlighting the need for an in vitro model of human adipocyte development. Here, we report a procedure to selectively derive BAP and WAPs from human-induced pluripotent stem cells. Molecular characterization of APs of both phenotypes revealed that BMP4, Hox8, Hoxc9, and HoxA5 genes were specifically expressed in WAPs, whereas expression of PRDM16, Dio2, and Pax3 marked BAPs. We focused on Pax3 and we showed that expression of this transcription factor was enriched in human perirenal white adipose tissue samples expressing UCP1 and in human classical brown fat. Finally, functional experiments indicated that Pax3 was a critical player of human AP fate as its ectopic expression led to convert WAPs into brown-like APs. Together, these data support a model in which Pax3 is a new marker of human BAPs and a molecular mediator of their fate. The findings of this study could lead to new anti-obesity therapies based on the recruitment of APs and constitute a platform for investigating in vitro the developmental origins of human white and brown adipocytes.
Collapse
Affiliation(s)
- Tala Mohsen-Kanson
- Université Nice Sophia Antipolis, iBV, UMR CNRS/INSERM, Faculté de Médecine, Nice Cedex, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Mônico-Neto M, Giampá SQDC, Lee KS, de Melo CM, Souza HDS, Dáttilo M, Minali PA, Santos Prado PH, Tufik S, de Mello MT, Antunes HKM. Negative energy balance induced by paradoxical sleep deprivation causes multicompartmental changes in adipose tissue and skeletal muscle. Int J Endocrinol 2015; 2015:908159. [PMID: 25821467 PMCID: PMC4364052 DOI: 10.1155/2015/908159] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 12/02/2022] Open
Abstract
Objective. Describe multicompartmental changes in the fat and various muscle fiber types, as well as the hormonal profile and metabolic rate induced by SD in rats. Methods. Twenty adult male Wistar rats were equally distributed into two groups: experimental group (EG) and control group (CG). The EG was submitted to SD for 96 h. Blood levels of corticosterone (CORT), total testosterone (TESTO), insulin like growth factor-1 (IGF-1), and thyroid hormones (T3 and T4) were used to assess the catabolic environment. Muscle trophism was measured using a cross-sectional area of various muscles (glycolytic, mixed, and oxidative), and lipolysis was inferred by the weight of fat depots from various locations, such as subcutaneous, retroperitoneal, and epididymal. The metabolic rate was measured using oxygen consumption ([Formula: see text]O2) measurement. Results. SD increased CORT levels and decreased TESTO, IGF-1, and T4. All fat depots were reduced in weight after SD. Glycolytic and mixed muscles showed atrophy, whereas atrophy was not observed in oxidative muscle. Conclusion. Our data suggest that glycolytic muscle fibers are more sensitive to atrophy than oxidative fibers during SD and that fat depots are reduced regardless of their location.
Collapse
Affiliation(s)
- Marcos Mônico-Neto
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros 925, 04024-003 São Paulo, SP, Brazil
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
| | - Sara Quaglia de Campos Giampá
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
- Departamento de Biociências, Universidade Federal de São Paulo, Rua Silva Jardim 136, 11015-020 Santos, SP, Brazil
| | - Kil Sun Lee
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, 04039-032 São Paulo, SP, Brazil
| | - Camila Maria de Melo
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros 925, 04024-003 São Paulo, SP, Brazil
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
| | - Helton de Sá Souza
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros 925, 04024-003 São Paulo, SP, Brazil
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
| | - Murilo Dáttilo
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros 925, 04024-003 São Paulo, SP, Brazil
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
| | - Paulo Alexandre Minali
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros 925, 04024-003 São Paulo, SP, Brazil
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
| | - Pedro Henrique Santos Prado
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
- Departamento de Biociências, Universidade Federal de São Paulo, Rua Silva Jardim 136, 11015-020 Santos, SP, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros 925, 04024-003 São Paulo, SP, Brazil
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
| | - Marco Túlio de Mello
- Departamento de Psicobiologia, Universidade Federal de São Paulo, Rua Napoleão de Barros 925, 04024-003 São Paulo, SP, Brazil
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
| | - Hanna Karen Moreira Antunes
- Centro de Estudos em Psicobiologia e Exercício, Rua Marselhesa 500, 04020-060 São Paulo, SP, Brazil
- Departamento de Biociências, Universidade Federal de São Paulo, Rua Silva Jardim 136, 11015-020 Santos, SP, Brazil
- *Hanna Karen Moreira Antunes:
| |
Collapse
|
69
|
De Sousa M, Porras DP, Perry CGR, Seale P, Scimè A. p107 is a crucial regulator for determining the adipocyte lineage fate choices of stem cells. Stem Cells 2014; 32:1323-36. [PMID: 24449206 DOI: 10.1002/stem.1637] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 11/20/2013] [Indexed: 12/11/2022]
Abstract
Thermogenic (beige and brown) adipocytes protect animals against obesity and metabolic disease. However, little is known about the mechanisms that commit stem cells toward different adipocyte lineages. We show here that p107 is a master regulator of adipocyte lineage fates, its suppression required for commitment of stem cells to the brown-type fate. p107 is strictly expressed in the stem cell compartment of white adipose tissue depots and completely absent in brown adipose tissue. Remarkably, p107-deficient stem cells uniformly give rise to brown-type adipocytes in vitro and in vivo. Furthermore, brown fat programming of mesenchymal stem cells by PRDM-BF1-RIZ1 homologous domain containing 16 (Prdm16) was associated with a dramatic reduction of p107 levels. Indeed, Prdm16 directly suppressed p107 transcription via promoter binding. Notably, the sustained expression of p107 blocked the ability of Prdm16 to induce brown fat genes. These findings demonstrate that p107 expression in stem cells commits cells to the white versus brown adipose lineage.
Collapse
Affiliation(s)
- Martina De Sousa
- Stem Cell Research Group, Faculty of Health, York University, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
70
|
Adipose tissue dysregulation and metabolic consequences in childhood and adolescent obesity: potential impact of dietary fat quality. Proc Nutr Soc 2014; 74:67-82. [PMID: 25497038 DOI: 10.1017/s002966511400158x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Evidence suggests that at a population level, childhood and adolescent obesity increase the long-term risk of chronic diseases such as type 2 diabetes and CVD. At an individual level, however, the metabolic consequences of obesity in youth vary immensely. Despite comparable BMI, some adolescents develop impaired glucose tolerance while others maintain normal glucose homeostasis. It has been proposed that the variation in the capacity to store lipid in the subcutaneous adipose tissue (SAT) may partially discriminate metabolically healthy from unhealthy obesity. In positive energy balance, a decreased capacity to expand SAT may drive lipid accumulation to visceral adipose tissue, liver and skeletal muscle. This state of lipotoxicity is associated with chronic low-grade inflammation, insulin resistance and dyslipidaemia. The present review examines the differential adipose tissue development and function in children and adolescents who exhibit metabolic dysregulation compared with those who are protected. Additionally, the role of manipulating dietary fat quality to potentially prevent and treat metabolic dysfunction in obesity will be discussed. The findings of the present review highlight the need for further randomised controlled trials to establish the effect of dietary n-3 PUFA on the metabolic phenotype of obese children and adolescents. Furthermore, using a personalised nutrition approach to target interventions to those at risk of, or those with established metabolic dysregulation may optimise the efficacy of modifying dietary fat quality.
Collapse
|
71
|
Rowland LA, Bal NC, Periasamy M. The role of skeletal-muscle-based thermogenic mechanisms in vertebrate endothermy. Biol Rev Camb Philos Soc 2014; 90:1279-97. [PMID: 25424279 DOI: 10.1111/brv.12157] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 10/03/2014] [Accepted: 10/14/2014] [Indexed: 12/17/2022]
Abstract
Thermogenesis is one of the most important homeostatic mechanisms that evolved during vertebrate evolution. Despite its importance for the survival of the organism, the mechanistic details behind various thermogenic processes remain incompletely understood. Although heat production from muscle has long been recognized as a thermogenic mechanism, whether muscle can produce heat independently of contraction remains controversial. Studies in birds and mammals suggest that skeletal muscle can be an important site of non-shivering thermogenesis (NST) and can be recruited during cold adaptation, although unequivocal evidence is lacking. Much research on thermogenesis during the last two decades has been focused on brown adipose tissue (BAT). These studies clearly implicate BAT as an important site of NST in mammals, in particular in newborns and rodents. However, BAT is either absent, as in birds and pigs, or is only a minor component, as in adult large mammals including humans, bringing into question the BAT-centric view of thermogenesis. This review focuses on the evolution and emergence of various thermogenic mechanisms in vertebrates from fish to man. A careful analysis of the existing data reveals that muscle was the earliest facultative thermogenic organ to emerge in vertebrates, long before the appearance of BAT in eutherian mammals. Additionally, these studies suggest that muscle-based thermogenesis is the dominant mechanism of heat production in many species including birds, marsupials, and certain mammals where BAT-mediated thermogenesis is absent or limited. We discuss the relevance of our recent findings showing that uncoupling of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA) by sarcolipin (SLN), resulting in futile cycling and increased heat production, could be the basis for NST in skeletal muscle. The overall goal of this review is to highlight the role of skeletal muscle as a thermogenic organ and provide a balanced view of thermogenesis in vertebrates.
Collapse
Affiliation(s)
- Leslie A Rowland
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Naresh C Bal
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, U.S.A
| |
Collapse
|
72
|
Hausman GJ, Basu U, Du M, Fernyhough-Culver M, Dodson MV. Intermuscular and intramuscular adipose tissues: Bad vs. good adipose tissues. Adipocyte 2014; 3:242-55. [PMID: 26317048 DOI: 10.4161/adip.28546] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/11/2014] [Accepted: 03/14/2014] [Indexed: 12/23/2022] Open
Abstract
Human studies of the influence of aging and other factors on intermuscular fat (INTMF) were reviewed. Intermuscular fat increased with weight loss, weight gain, or with no weight change with age in humans. An increase in INTMF represents a similar threat to type 2 diabetes and insulin resistance as does visceral adipose tissue (VAT). Studies of INTMF in animals covered topics such as quantitative deposition and genetic relationships with other fat depots. The relationship between leanness and higher proportions of INTMF fat in pigs was not observed in human studies and was not corroborated by other pig studies. In humans, changes in muscle mass, strength and quality are associated with INTMF accretion with aging. Gene expression profiling and intrinsic methylation differences in pigs demonstrated that INTMF and VAT are primarily associated with inflammatory and immune processes. It seems that in the pig and humans, INTMF and VAT share a similar pattern of distribution and a similar association of components dictating insulin sensitivity. Studies on intramuscular (IM) adipocyte development in meat animals were reviewed. Gene expression analysis and genetic analysis have identified candidate genes involved in IM adipocyte development. Intramuscular (IM) adipocyte development in human muscle is only seen during aging and some pathological circumstance. Several genetic links between human and meat animal adipogenesis have been identified. In pigs, the Lipin1 and Lipin 2 gene have strong genetic effects on IM accumulation. Lipin1 deficiency results in immature adipocyte development in human lipodystrophy. In humans, overexpression of Perilipin 2 (PLIN2) facilitates intramyocellular lipid accretion whereas in pigs PLIN2 gene expression is associated with IM deposition. Lipins and perilipins may influence intramuscular lipid regardless of species.
Collapse
|
73
|
Hafner AL, Dani C. Human induced pluripotent stem cells: A new source for brown and white adipocytes. World J Stem Cells 2014; 6:467-472. [PMID: 25258668 PMCID: PMC4172675 DOI: 10.4252/wjsc.v6.i4.467] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/25/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) derived from human induced pluripotent stem cells (hiPSCs) provide a novel source for generating adipocytes, thus opening new avenues for fundamental research and clinical medicine. We present the adipogenic potential of hiPSCs and the various methods to derive hiPSC-MSCs. We discuss the main characteristic of hiPSC-MSCs, which is their low adipogenic capacity as compared to adult-MSCs. Finally, we propose several hypotheses to explanation this feature, underlying a potential critical role of the micro-environment. We favour the hypothesis that the range of factors or culture conditions required to induce adipocyte differentiation of MSCs derived from adult tissues and from embryonic-like cells could differ.
Collapse
|
74
|
Gunawardana SC. Benefits of healthy adipose tissue in the treatment of diabetes. World J Diabetes 2014; 5:420-430. [PMID: 25126390 PMCID: PMC4127579 DOI: 10.4239/wjd.v5.i4.420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/24/2014] [Accepted: 06/03/2014] [Indexed: 02/05/2023] Open
Abstract
The major malfunction in diabetes mellitus is severe perturbation of glucose homeostasis caused by deficiency of insulin. Insulin deficiency is either absolute due to destruction or failure of pancreatic β cells, or relative due to decreased sensitivity of peripheral tissues to insulin. The primary lesion being related to insulin, treatments for diabetes focus on insulin replacement and/or increasing sensitivity to insulin. These therapies have their own limitations and complications, some of which can be life-threatening. For example, exogenous insulin administration can lead to fatal hypoglycemic episodes; islet/pancreas transplantation requires life-long immunosuppressive therapy; and anti-diabetic drugs have dangerous side effects including edema, heart failure and lactic acidosis. Thus the need remains for better safer long term treatments for diabetes. The ultimate goal in treating diabetes is to re-establish glucose homeostasis, preferably through endogenously generated hormones. Recent studies increasingly show that extra-pancreatic hormones, particularly those arising from adipose tissue, can compensate for insulin, or entirely replace the function of insulin under appropriate circumstances. Adipose tissue is a versatile endocrine organ that secretes a variety of hormones with far-reaching effects on overall metabolism. While unhealthy adipose tissue can exacerbate diabetes through limiting circulation and secreting of pro-inflammatory cytokines, healthy uninflamed adipose tissue secretes beneficial adipokines with hypoglycemic and anti-inflammatory properties, which can complement and/or compensate for the function of insulin. Administration of specific adipokines is known to alleviate both type 1 and 2 diabetes, and leptin mono-therapy is reported to reverse type 1 diabetes independent of insulin. Although specific adipokines may correct diabetes, administration of individual adipokines still carries risks similar to those of insulin monotherapy. Thus a better approach is to achieve glucose homeostasis with endogenously-generated adipokines through transplantation or regeneration of healthy adipose tissue. Our recent studies on mouse models show that type 1 diabetes can be reversed without insulin through subcutaneous transplantation of embryonic brown adipose tissue, which leads to replenishment of recipients’ white adipose tissue; increase of a number of beneficial adipokines; and fast and long-lasting euglycemia. Insulin-independent glucose homeostasis is established through a combination of endogenously generated hormones arising from the transplant and/or newly-replenished white adipose tissue. Transplantation of healthy white adipose tissue is reported to alleviate type 2 diabetes in rodent models on several occasions, and increasing the content of endogenous brown adipose tissue is known to combat obesity and type 2 diabetes in both humans and animal models. While the underlying mechanisms are not fully documented, the beneficial effects of healthy adipose tissue in improving metabolism are increasingly reported, and are worthy of attention as a powerful tool in combating metabolic disease.
Collapse
|
75
|
Hudak CS, Gulyaeva O, Wang Y, Park SM, Lee L, Kang C, Sul HS. Pref-1 marks very early mesenchymal precursors required for adipose tissue development and expansion. Cell Rep 2014; 8:678-87. [PMID: 25088414 DOI: 10.1016/j.celrep.2014.06.060] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 05/07/2014] [Accepted: 06/26/2014] [Indexed: 11/19/2022] Open
Abstract
Pref-1 is an EGF-repeat-containing protein that inhibits adipocyte differentiation. To better understand the origin and development of white adipose tissue (WAT), we generated transgenic mouse models for transient or permanent fluorescent labeling of cells using the Pref-1 promoter, facilitating inducible ablation. We show that Pref-1-marked cells retain proliferative capacity and are very early adipose precursors, prior to expression of Zfp423 or PPARγ. In addition, the Pref-1-marked cells establish that adipose precursors are mesenchymal, but not endothelial or pericytal, in origin. During embryogenesis, Pref-1-marked cells first appear in the dorsal mesenteric region as early as embryonic day 10.5 (E10.5). These cells become lipid-laden adipocytes at E17.5 in the subcutaneous region, whereas visceral WAT develops after birth. Finally, ablation of Pref-1-marked cells prevents not only embryonic WAT development but also later adult adipose expansion upon high-fat feeding, demonstrating the requirement of Pref-1 cells for adipogenesis.
Collapse
Affiliation(s)
- Carolyn S Hudak
- Department of Nutritional Sciences & Toxicology, 119 Morgan Hall, University of California, Berkeley, CA 94720, USA
| | - Olga Gulyaeva
- Endocrinology Program, 299 LSA, University of California, Berkeley, CA 94720, USA
| | - Yuhui Wang
- Department of Nutritional Sciences & Toxicology, 119 Morgan Hall, University of California, Berkeley, CA 94720, USA
| | - Seung-Min Park
- Department of Bioengineering, 306 Stanley Hall, University of California, Berkeley, CA 94720 USA; Berkeley Sensor and Actuator Center, 403 Cory Hall, University of California, Berkeley, CA 94720, USA; Department of Radiology, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Luke Lee
- Department of Bioengineering, 306 Stanley Hall, University of California, Berkeley, CA 94720 USA
| | - Chulho Kang
- Department of Molecular & Cell Biology, 142 LSA, University of California, Berkeley, CA 94720, USA
| | - Hei Sook Sul
- Department of Nutritional Sciences & Toxicology, 119 Morgan Hall, University of California, Berkeley, CA 94720, USA; Endocrinology Program, 299 LSA, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
76
|
Abstract
Cell-lineage tracing has revealed a complex heterogeneity present in postnatal tissue and adult progenitors. Chau et al. (2014) and Long et al. (2014) provide further evidence for this among adipocytes, and their findings underscore the importance of cellular ontogeny not just for development but also for potential treatment of disease.
Collapse
|
77
|
Khalyfa A, Wang Y, Zhang SX, Qiao Z, Abdelkarim A, Gozal D. Sleep fragmentation in mice induces nicotinamide adenine dinucleotide phosphate oxidase 2-dependent mobilization, proliferation, and differentiation of adipocyte progenitors in visceral white adipose tissue. Sleep 2014; 37:999-1009. [PMID: 24790279 DOI: 10.5665/sleep.3678] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chronic sleep fragmentation (SF) without sleep curtailment induces increased adiposity. However, it remains unclear whether mobilization, proliferation, and differentiation of adipocyte progenitors (APs) occurs in visceral white adipose tissue (VWAT), and whether nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2) activity plays a role. METHODS Changes in VWAT depot cell size and AP proliferation were assessed in wild-type and Nox2 null male mice exposed to SF and control sleep (SC). To assess mobilization, proliferation, and differentiation of bone marrow mesenchymal stem cells (BM-MSC), Sca-1+ bone marrow progenitors were isolated from GFP+ or RFP+ mice, and injected intravenously to adult male mice (C57BL/6) previously exposed to SF or SC. RESULTS In comparison with SC, SF was associated with increased weight accrual at 3 w and thereafter, larger subcutaneous and visceral fat depots, and overall adipocyte size at 8 w. Increased global AP numbers in VWAT along with enhanced AP BrDU labeling in vitro and in vivo emerged in SF. Systemic injections of GFP+ BM-MSC resulted in increased AP in VWAT, as well as in enhanced differentiation into adipocytes in SF-exposed mice. No differences occurred between SF and SC in Nox2 null mice for any of these measurements. CONCLUSIONS Chronic sleep fragmentation (SF) induces obesity in mice and increased proliferation and differentiation of adipocyte progenitors (AP) in visceral white adipose tissue (VWAT) that are mediated by increased Nox2 activity. In addition, enhanced migration of bone marrow mesenchymal stem cells from the systemic circulation into VWAT, along with AP differentiation, proliferation, and adipocyte formation occur in SF-exposed wild-type but not in oxidase 2 (Nox2) null mice. Thus, Nox2 may provide a therapeutic target to prevent obesity in the context of sleep disorders.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Shelley X Zhang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Zhuanhong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Amal Abdelkarim
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|
78
|
Tarantal AF, Berglund L. Obesity and lifespan health--importance of the fetal environment. Nutrients 2014; 6:1725-36. [PMID: 24763115 PMCID: PMC4011063 DOI: 10.3390/nu6041725] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/09/2014] [Accepted: 04/15/2014] [Indexed: 02/02/2023] Open
Abstract
A marked increase in the frequency of obesity at the population level has resulted in an increasing number of obese women entering pregnancy. The increasing realization of the importance of the fetal environment in relation to chronic disease across the lifespan has focused attention on the role of maternal obesity in fetal development. Previous studies have demonstrated that obesity during adolescence and adulthood can be traced back to fetal and early childhood exposures. This review focuses on factors that contribute to early developmental events, such as epigenetic modifications, the potential for an increase in inflammatory burden, early developmental programming changes such as the variable development of white versus brown adipose tissue, and alterations in organ ontogeny. We hypothesize that these mechanisms promote an unfavorable fetal environment and can have a long-standing impact, with early manifestations of chronic disease that can result in an increased demand for future health care. In order to identify appropriate preventive measures, attention needs to be placed both on reducing maternal obesity as well as understanding the molecular, cellular, and epigenetic mechanisms that may be responsible for the prenatal onset of chronic disease.
Collapse
Affiliation(s)
- Alice F Tarantal
- Department of Pediatrics, School of Medicine, University of California, Davis, CA 95616, USA.
| | - Lars Berglund
- Department of Medicine, School of Medicine, University of California, Davis, CA 95616, USA.
| |
Collapse
|
79
|
Chau YY, Bandiera R, Serrels A, Martínez-Estrada OM, Qing W, Lee M, Slight J, Thornburn A, Berry R, McHaffie S, Stimson RH, Walker BR, Chapuli RM, Schedl A, Hastie N. Visceral and subcutaneous fat have different origins and evidence supports a mesothelial source. Nat Cell Biol 2014; 16:367-75. [PMID: 24609269 PMCID: PMC4060514 DOI: 10.1038/ncb2922] [Citation(s) in RCA: 384] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 01/27/2014] [Indexed: 12/14/2022]
Abstract
Fuelled by the obesity epidemic, there is considerable interest in the developmental origins of white adipose tissue (WAT) and the stem and progenitor cells from which it arises. Whereas increased visceral fat mass is associated with metabolic dysfunction, increased subcutaneous WAT is protective. There are six visceral fat depots: perirenal, gonadal, epicardial, retroperitoneal, omental and mesenteric, and it is a subject of much debate whether these have a common developmental origin and whether this differs from that for subcutaneous WAT. Here we show that all six visceral WAT depots receive a significant contribution from cells expressing Wt1 late in gestation. Conversely, no subcutaneous WAT or brown adipose tissue arises from Wt1-expressing cells. Postnatally, a subset of visceral WAT continues to arise from Wt1-expressing cells, consistent with the finding that Wt1 marks a proportion of cell populations enriched in WAT progenitors. We show that all visceral fat depots have a mesothelial layer like the visceral organs with which they are associated, and provide several lines of evidence that Wt1-expressing mesothelium can produce adipocytes. These results reveal a major ontogenetic difference between visceral and subcutaneous WAT, and pinpoint the lateral plate mesoderm as a major source of visceral WAT. They also support the notion that visceral WAT progenitors are heterogeneous, and suggest that mesothelium is a source of adipocytes.
Collapse
Affiliation(s)
- You-Ying Chau
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Roberto Bandiera
- IBV, INSERM U1091, Univeriste de Nice Sophia-Antipolis, Parc Valrose, Centre de Biochimie, 06100 Nice Cedex-2 FRANCE
| | - Alan Serrels
- Institute for Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh Cancer Research UK Centre, Western General Hospital Campus, Crewe Road South, Edinburgh, EH4 2XR
| | - Ofelia M Martínez-Estrada
- Department of Cell Biology, Faculty of Biology, University of Barcelona, Av. Diagonal, 643, 08028 Barcelona, Spain
| | - Wei Qing
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Martin Lee
- Institute for Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh Cancer Research UK Centre, Western General Hospital Campus, Crewe Road South, Edinburgh, EH4 2XR
| | - Joan Slight
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Anna Thornburn
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Rachel Berry
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Sophie McHaffie
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Roland H Stimson
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | - Andreas Schedl
- IBV, INSERM U1091, Univeriste de Nice Sophia-Antipolis, Parc Valrose, Centre de Biochimie, 06100 Nice Cedex-2 FRANCE
| | - Nick Hastie
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| |
Collapse
|
80
|
Sanchez-Gurmaches J, Guertin DA. Adipocyte lineages: tracing back the origins of fat. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:340-51. [PMID: 23747579 PMCID: PMC3805734 DOI: 10.1016/j.bbadis.2013.05.027] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 12/25/2022]
Abstract
The obesity epidemic has intensified efforts to understand the mechanisms controlling adipose tissue development. Adipose tissue is generally classified as white adipose tissue (WAT), the major energy storing tissue, or brown adipose tissue (BAT), which mediates non-shivering thermogenesis. It is hypothesized that brite adipocytes (brown in white) may represent a third adipocyte class. The recent realization that brown fat exist in adult humans suggests increasing brown fat energy expenditure could be a therapeutic strategy to combat obesity. To understand adipose tissue development, several groups are tracing the origins of mature adipocytes back to their adult precursor and embryonic ancestors. From these studies emerged a model that brown adipocytes originate from a precursor shared with skeletal muscle that expresses Myf5-Cre, while all white adipocytes originate from a Myf5-negative precursors. While this provided a rational explanation to why BAT is more metabolically favorable than WAT, recent work indicates the situation is more complex because subsets of white adipocytes also arise from Myf5-Cre expressing precursors. Lineage tracing studies further suggest that the vasculature may provide a niche supporting both brown and white adipocyte progenitors; however, the identity of the adipocyte progenitor cell is under debate. Differences in origin between adipocytes could explain metabolic heterogeneity between depots and/or influence body fat patterning particularly in lipodystrophy disorders. Here, we discuss recent insights into adipose tissue origins highlighting lineage-tracing studies in mice, how variations in metabolism or signaling between lineages could affect body fat distribution, and the questions that remain unresolved. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
81
|
Zhang Y, Zitsman JL, Hou J, Fennoy I, Guo K, Feinberg J, Leibel RL. Fat cell size and adipokine expression in relation to gender, depot, and metabolic risk factors in morbidly obese adolescents. Obesity (Silver Spring) 2014; 22:691-7. [PMID: 23804589 PMCID: PMC3823663 DOI: 10.1002/oby.20528] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 06/04/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To understand the regulation of adipocyte size and adipokine expression in relation to gender, anatomic location, adiposity, and metabolic risk factors in adolescents with morbid obesity. METHODS Adipocyte size and adipokine expression in paired abdominal subcutaneous (SAT) and omental (VAT) surgical adipose tissues were related to gender, anatomic location, adiposity, and metabolic risk factors in a group of morbidly obese adolescents. RESULTS Significant depot- and/or gender-related differences in adipocyte size and adipokine expression were detected. Adjusted for body mass index, adipocyte size in both depots was larger in males than in females and was a major predictor of mRNA levels of leptin, plasminogen activator inhibitor-1, and adiponectin. Gender, but not adipocyte size, was significantly correlated with proinflammatory cytokine expression. Body mass index and waist circumference were correlated positively with VAT adipocyte size and negatively with SAT adipocyte size. VAT adiponectin and interleukin-6 expression levels were major predictors of high-density lipoprotein cholesterol concentrations, independent of gender, adiposity, and insulin sensitivity. CONCLUSIONS Adipose tissue morphology and function in obese adolescents are influenced by gender and anatomic location; the pattern of gender- and depot-related differences in adipocyte size and adipokine expression suggests that adolescent males, relative to the females, are at increased risk for obesity-related metabolic comorbidities.
Collapse
Affiliation(s)
- Yiying Zhang
- Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, USA; Naomi Berrie Diabetes Center, Columbia University, College of Physicians and Surgeons, New York, USA
| | | | | | | | | | | | | |
Collapse
|
82
|
Carrière A, Jeanson Y, Cousin B, Arnaud E, Casteilla L. Le recrutement et l’activation d’adipocytes bruns et/ou BRITE. Med Sci (Paris) 2013; 29:729-35. [DOI: 10.1051/medsci/2013298011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
83
|
Ancestral Myf5 gene activity in periocular connective tissue identifies a subset of fibro/adipogenic progenitors but does not connote a myogenic origin. Dev Biol 2013; 385:366-79. [PMID: 23969310 DOI: 10.1016/j.ydbio.2013.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/21/2013] [Accepted: 08/13/2013] [Indexed: 11/23/2022]
Abstract
Extraocular muscles (EOM) represent a unique muscle group that controls eye movements and originates from head mesoderm, while the more typically studied body and limb muscles are somite-derived. Aiming to investigate myogenic progenitors (satellite cells) in EOM versus limb and diaphragm of adult mice, we have been using flow cytometry in combination with myogenic-specific Cre-loxP lineage marking for cell isolation. While analyzing cells from the EOM of mice that harbor Myf5(Cre)-driven GFP expression, we identified in addition to the expected GFP(+) myogenic cells (presumably satellite cells), a second dominant GFP(+) population distinguished as being Sca1(+), non-myogenic, and exhibiting a fibro/adipogenic potential. This unexpected population was not only unique to EOM compared to the other muscles but also specific to the Myf5(Cre)-driven reporter when compared to the MyoD(Cre) driver. Histological studies of periocular tissue preparations demonstrated the presence of Myf5(Cre)-driven GFP(+) cells in connective tissue locations adjacent to the muscle masses, including cells in the vasculature wall. These vasculature-associated GFP(+) cells were further identified as mural cells based on the presence of the specific XLacZ4 transgene. Unlike the EOM satellite cells that originate from a Pax3-negative lineage, these non-myogenic Myf5(Cre)-driven GFP(+) cells appear to be related to cells of a Pax3-expressing origin, presumably derived from the neural crest. In all, our lineage tracing based on multiple reporter lines has demonstrated that regardless of common ancestral expression of Myf5, there is a clear distinction between periocular myogenic and non-myogenic cell lineages according to their mutually exclusive antecedence of MyoD and Pax3 gene activity.
Collapse
|
84
|
Gerhard GS, Styer AM, Strodel WE, Roesch SL, Yavorek A, Carey DJ, Wood GC, Petrick AT, Gabrielsen J, Ibele A, Benotti P, Rolston DD, Still CD, Argyropoulos G. Gene expression profiling in subcutaneous, visceral and epigastric adipose tissues of patients with extreme obesity. Int J Obes (Lond) 2013; 38:371-8. [PMID: 23949615 PMCID: PMC3925764 DOI: 10.1038/ijo.2013.152] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 07/19/2013] [Accepted: 08/08/2013] [Indexed: 02/08/2023]
Abstract
Objective The goal of the present study was to identify differences in gene expression between SAT, VAT, and EAT depots in Class III severely obese individuals. Design Human subcutaneous (SAT) and visceral (VAT) adipose tissues exhibit differential gene expression profiles. There is little information, however, about the other proximal white adipose tissue, epigastric (EAT) in terms of its function and contribution to metabolism. Subjects and Methods Using RNA from adipose biospecimens obtained from Class III severely obese patients undergoing open Roux-en-Y gastric bypass surgery, we compared gene expression profiles between SAT, VAT, and EAT, using microarrays validated by real time quantitative PCR. Results The three depots were found to share 1,907 genes. VAT had the greatest number of genes [66] expressed exclusively in this depot, followed by SAT [23], and then EAT [14]. Moreover, VAT shared more genes with EAT [65] than with SAT [38]. Further analyses using ratios of SAT/EAT, VAT/EAT, and SAT/VAT, identified specific as well as overlapping networks and pathways of genes representing dermatological diseases, inflammation, cell cycle and growth, cancer, and development. Targeted analysis of genes playing a role in adipose tissue development and function, revealed that Peroxisome proliferator-activated receptor Gamma Coactivator 1-alpha (PGC1-α) that regulates the precursor of the hormone Irisin (FNCD5), were abundantly expressed in all three fat depots, along with fibroblast growth factors (FGF) FGF1, FGF7, and FGF10, whereas, FGF19 and FGF21 were undetectable. Conclusions These data indicate that EAT has more in common with VAT suggesting similar metabolic potential. The human epigastric adipose depot could play a significant functional role in metabolic diseases and should be further investigated.
Collapse
Affiliation(s)
- G S Gerhard
- 1] Weis Center for Research, Danville, PA, USA [2] Department of Biochemistry and Molecular Biology and Department of Pathology and Laboratory Medicine, Pennsylvania State University, Hershey, PA, USA
| | - A M Styer
- Weis Center for Research, Danville, PA, USA
| | - W E Strodel
- Department of Surgery, Geisinger Medical Center, Danville, PA, USA
| | - S L Roesch
- Weis Center for Research, Danville, PA, USA
| | - A Yavorek
- Weis Center for Research, Danville, PA, USA
| | - D J Carey
- Weis Center for Research, Danville, PA, USA
| | - G C Wood
- Institute of Obesity, Geisinger Medical Center, Danville, PA, USA
| | - A T Petrick
- Department of Surgery, Geisinger Medical Center, Danville, PA, USA
| | - J Gabrielsen
- Department of Surgery, Geisinger Medical Center, Danville, PA, USA
| | - A Ibele
- Department of Surgery, Geisinger Medical Center, Danville, PA, USA
| | - P Benotti
- Institute of Obesity, Geisinger Medical Center, Danville, PA, USA
| | - D D Rolston
- Department of General and Internal Medicine, Geisinger Medical Center, Danville, PA, USA
| | - C D Still
- 1] Institute of Obesity, Geisinger Medical Center, Danville, PA, USA [2] Department of Gastroenterology, Geisinger Medical Center, Danville, PA, USA
| | - G Argyropoulos
- 1] Weis Center for Research, Danville, PA, USA [2] Institute of Obesity, Geisinger Medical Center, Danville, PA, USA
| |
Collapse
|
85
|
Mueller E. Understanding the variegation of fat: novel regulators of adipocyte differentiation and fat tissue biology. Biochim Biophys Acta Mol Basis Dis 2013; 1842:352-7. [PMID: 23735215 DOI: 10.1016/j.bbadis.2013.05.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 01/06/2023]
Abstract
The differentiation of uncommitted cells into specialized adipocytes occurs through a cascade of transcriptional events culminating in the induction and activation of the nuclear receptor PPARγ, the central coordinator of fat cell function. Since the discovery of PPARγ, two decades ago, our views of how this molecule is activated have been significantly refined. Beyond the cell, we also now know that diverse signals and regulators control PPARγ function in a fat-depot specific manner. The goal of this article is to review the latest in our understanding of the early and late transcriptional events that regulate adipocyte development and their potential impact on energy storage and expenditure in different fat depots. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Elisabetta Mueller
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
86
|
Lee YH, Mottillo EP, Granneman JG. Adipose tissue plasticity from WAT to BAT and in between. Biochim Biophys Acta Mol Basis Dis 2013; 1842:358-69. [PMID: 23688783 DOI: 10.1016/j.bbadis.2013.05.011] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/21/2013] [Accepted: 05/06/2013] [Indexed: 01/09/2023]
Abstract
Adipose tissue plays an essential role in regulating energy balance through its metabolic, cellular and endocrine functions. Adipose tissue has been historically classified into anabolic white adipose tissue and catabolic brown adipose tissue. An explosion of new data, however, points to the remarkable heterogeneity among the cells types that can become adipocytes, as well as the inherent metabolic plasticity of mature cells. These data indicate that targeting cellular and metabolic plasticity of adipose tissue might provide new avenues for treatment of obesity-related diseases. This review will discuss the developmental origins of adipose tissue, the cellular complexity of adipose tissues, and the identification of progenitors that contribute to adipogenesis throughout development. We will touch upon the pathological remodeling of adipose tissue and discuss how our understanding of adipose tissue remodeling can uncover new therapeutic targets. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Yun-Hee Lee
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Emilio P Mottillo
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - James G Granneman
- Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
87
|
Wojciechowicz K, Gledhill K, Ambler CA, Manning CB, Jahoda CAB. Development of the mouse dermal adipose layer occurs independently of subcutaneous adipose tissue and is marked by restricted early expression of FABP4. PLoS One 2013; 8:e59811. [PMID: 23555789 PMCID: PMC3608551 DOI: 10.1371/journal.pone.0059811] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 02/18/2013] [Indexed: 12/05/2022] Open
Abstract
The laboratory mouse is a key animal model for studies of adipose biology, metabolism and disease, yet the developmental changes that occur in tissues and cells that become the adipose layer in mouse skin have received little attention. Moreover, the terminology around this adipose body is often confusing, as frequently no distinction is made between adipose tissue within the skin, and so called subcutaneous fat. Here adipocyte development in mouse dorsal skin was investigated from before birth to the end of the first hair follicle growth cycle. Using Oil Red O staining, immunohistochemistry, quantitative RT-PCR and TUNEL staining we confirmed previous observations of a close spatio-temporal link between hair follicle development and the process of adipogenesis. However, unlike previous studies, we observed that the skin adipose layer was created from cells within the lower dermis. By day 16 of embryonic development (e16) the lower dermis was demarcated from the upper dermal layer, and commitment to adipogenesis in the lower dermis was signalled by expression of FABP4, a marker of adipocyte differentiation. In mature mice the skin adipose layer is separated from underlying subcutaneous adipose tissue by the panniculus carnosus. We observed that the skin adipose tissue did not combine or intermix with subcutaneous adipose tissue at any developmental time point. By transplanting skin isolated from e14.5 mice (prior to the start of adipogenesis), under the kidney capsule of adult mice, we showed that skin adipose tissue develops independently and without influence from subcutaneous depots. This study has reinforced the developmental link between hair follicles and skin adipocyte biology. We argue that because skin adipocytes develop from cells within the dermis and independently from subcutaneous adipose tissue, that it is accurately termed dermal adipose tissue and that, in laboratory mice at least, it represents a separate adipose depot.
Collapse
Affiliation(s)
- Kamila Wojciechowicz
- School of Biological and Biomedical Sciences, Durham University, Durham, County Durham, United Kingdom
- Biological Stress Response, The Netherlands Cancer Institute, Amsterdam, North Holland, The Netherlands
| | - Karl Gledhill
- School of Biological and Biomedical Sciences, Durham University, Durham, County Durham, United Kingdom
- * E-mail:
| | - Carrie A. Ambler
- School of Biological and Biomedical Sciences, Durham University, Durham, County Durham, United Kingdom
| | - Craig B. Manning
- School of Biological and Biomedical Sciences, Durham University, Durham, County Durham, United Kingdom
| | - Colin A. B. Jahoda
- School of Biological and Biomedical Sciences, Durham University, Durham, County Durham, United Kingdom
| |
Collapse
|
88
|
Modica S, Wolfrum C. Bone morphogenic proteins signaling in adipogenesis and energy homeostasis. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:915-23. [PMID: 23353598 DOI: 10.1016/j.bbalip.2013.01.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Revised: 01/07/2013] [Accepted: 01/10/2013] [Indexed: 01/07/2023]
Abstract
A great deal is known about the molecular mechanisms regulating terminal differentiation of pre-adipocytes into mature adipocytes. In contrast, the knowledge about pathways that trigger commitment of mesenchymal stem cells into the adipocyte lineage is fragmented. In recent years, the role of members of the bone morphogenic protein family in regulating the early steps of adipogenesis has been the focus of research. Findings based on these studies have also highlighted an unexpected role for some bone morphogenic protein in energy homeostasis via regulation of adipocyte development and function. This review summarizes the knowledge about bone morphogenic proteins and their role in adipocyte commitment and regulation of whole body energy homeostasis. This article is part of a Special Issue entitled Brown and White Fat: From Signaling to Disease.
Collapse
Affiliation(s)
- Salvatore Modica
- Institute of Food, Nutrition and Health, ETH Zurich, Switzerland
| | | |
Collapse
|
89
|
Bénézech C, Mader E, Desanti G, Khan M, Nakamura K, White A, Ware CF, Anderson G, Caamaño JH. Lymphotoxin-β receptor signaling through NF-κB2-RelB pathway reprograms adipocyte precursors as lymph node stromal cells. Immunity 2012; 37:721-34. [PMID: 22940098 PMCID: PMC3809035 DOI: 10.1016/j.immuni.2012.06.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 06/20/2012] [Accepted: 06/30/2012] [Indexed: 01/08/2023]
Abstract
Lymph node development during embryogenesis involves lymphotoxin-β receptor engagement and subsequent differentiation of a poorly defined population of mesenchymal cells into lymphoid tissue organizer cells. Here, we showed that embryonic mesenchymal cells with characteristics of adipocyte precursors present in the microenvironment of lymph nodes gave rise to lymph node organizer cells. Signaling through the lymphotoxin-β receptor controlled the fate of adipocyte precursor cells by blocking adipogenesis and instead promoting lymphoid tissue stromal cell differentiation. This effect involved activation of the NF-κB2-RelB signaling pathway and inhibition of the expression of the key adipogenic factors Pparγ and Cebpα. In vivo organogenesis assays show that embryonic and adult adipocyte precursor cells can migrate into newborn lymph nodes and differentiate into a variety of lymph node stromal cells. Thus, we propose that adipose tissues act as a source of lymphoid stroma for lymph nodes and other lymphoid structures associated with fat.
Collapse
Affiliation(s)
- Cécile Bénézech
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Emma Mader
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Guillaume Desanti
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Mahmood Khan
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Kyoko Nakamura
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Andrea White
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Carl F. Ware
- Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Graham Anderson
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jorge H. Caamaño
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
90
|
Gunawardana SC. Therapeutic value of brown adipose tissue: Correcting metabolic disease through generating healthy fat. Adipocyte 2012; 1:250-255. [PMID: 23700541 PMCID: PMC3609108 DOI: 10.4161/adip.21042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Brown adipose tissue (BAT), an important endocrine organ long known for thermogenesis and energy consumption, has received much attention in recent years for its potential to combat obesity. In general, BAT can enhance metabolism and improve overall health. Our recent work demonstrates the ability of embryonic BAT transplants to correct type 1 diabetes (T1D) without insulin, via mechanisms somewhat different from those involved in BAT-associated weight loss. BAT transplants seem to reverse T1D by decreasing inflammation and increasing functionality in the surrounding white adipose tissue (WAT), thereby enabling it to secrete hypoglycemic adipokines, which compensate for the function of insulin. Thus BAT can transform unhealthy WAT to a healthy status, sufficient to replace the function of endocrine pancreas and establish insulin-independent glycemic regulation. Several studies, including ours, demonstrate the remarkable ability of BAT to correct metabolic disorders and hint at its beneficial effects on inflammation. Hence, addition of more BAT to the body, through transplantation or stimulating regeneration, may well be the therapy of the future for the simple correction of numerous diseases.
Collapse
|
91
|
Sanchez-Gurmaches J, Hung CM, Sparks CA, Tang Y, Li H, Guertin DA. PTEN loss in the Myf5 lineage redistributes body fat and reveals subsets of white adipocytes that arise from Myf5 precursors. Cell Metab 2012; 16:348-62. [PMID: 22940198 PMCID: PMC3488151 DOI: 10.1016/j.cmet.2012.08.003] [Citation(s) in RCA: 263] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 03/16/2012] [Accepted: 08/09/2012] [Indexed: 02/07/2023]
Abstract
The developmental origin of adipose tissue and what controls its distribution is poorly understood. By lineage tracing and gene expression analysis in mice, we provide evidence that mesenchymal precursors expressing Myf5--which are thought to give rise only to brown adipocytes and skeletal muscle--also give rise to a subset of white adipocytes. Furthermore, individual brown and white fats contain a mixture of adipocyte progenitor cells derived from Myf5(+) and Myf5(neg) lineages, the number of which varies with depot location. Subsets of white adipocytes originating from both Myf5(+) and Myf5(neg) precursors respond to β(3)-adrenoreceptor stimulation, suggesting "brite" adipocytes may also have multiple origins. We additionally find that deleting PTEN with myf5-cre causes lipomatosis and partial lipodystrophy by selectively expanding the Myf5(+) adipocyte lineages. Thus, the spectrum of adipocytes arising from Myf5(+) precursors is broader than previously thought, and differences in PI3K activity between adipocyte lineages alter body fat distribution.
Collapse
Affiliation(s)
- Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | |
Collapse
|
92
|
Wu J, Boström P, Sparks LM, Ye L, Choi JH, Giang AH, Khandekar M, Virtanen KA, Nuutila P, Schaart G, Huang K, Tu H, van Marken Lichtenbelt WD, Hoeks J, Enerbäck S, Schrauwen P, Spiegelman BM. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell 2012; 150:366-76. [PMID: 22796012 DOI: 10.1016/j.cell.2012.05.016] [Citation(s) in RCA: 2489] [Impact Index Per Article: 207.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/03/2012] [Accepted: 05/07/2012] [Indexed: 12/11/2022]
Abstract
Brown fat generates heat via the mitochondrial uncoupling protein UCP1, defending against hypothermia and obesity. Recent data suggest that there are two distinct types of brown fat: classical brown fat derived from a myf-5 cellular lineage and UCP1-positive cells that emerge in white fat from a non-myf-5 lineage. Here, we report the isolation of "beige" cells from murine white fat depots. Beige cells resemble white fat cells in having extremely low basal expression of UCP1, but, like classical brown fat, they respond to cyclic AMP stimulation with high UCP1 expression and respiration rates. Beige cells have a gene expression pattern distinct from either white or brown fat and are preferentially sensitive to the polypeptide hormone irisin. Finally, we provide evidence that previously identified brown fat deposits in adult humans are composed of beige adipocytes. These data provide a foundation for studying this mammalian cell type with therapeutic potential. PAPERCLIP:
Collapse
Affiliation(s)
- Jun Wu
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Klingenspor M, Herzig S, Pfeifer A. Brown fat develops a brite future. Obes Facts 2012; 5:890-6. [PMID: 23296106 DOI: 10.1159/000346337] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 01/02/2023] Open
|