101
|
Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, Beguinot F, Miele C. Adipose Tissue Dysfunction as Determinant of Obesity-Associated Metabolic Complications. Int J Mol Sci 2019; 20:ijms20092358. [PMID: 31085992 PMCID: PMC6539070 DOI: 10.3390/ijms20092358] [Citation(s) in RCA: 983] [Impact Index Per Article: 163.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a critical risk factor for the development of type 2 diabetes (T2D), and its prevalence is rising worldwide. White adipose tissue (WAT) has a crucial role in regulating systemic energy homeostasis. Adipose tissue expands by a combination of an increase in adipocyte size (hypertrophy) and number (hyperplasia). The recruitment and differentiation of adipose precursor cells in the subcutaneous adipose tissue (SAT), rather than merely inflating the cells, would be protective from the obesity-associated metabolic complications. In metabolically unhealthy obesity, the storage capacity of SAT, the largest WAT depot, is limited, and further caloric overload leads to the fat accumulation in ectopic tissues (e.g., liver, skeletal muscle, and heart) and in the visceral adipose depots, an event commonly defined as “lipotoxicity.” Excessive ectopic lipid accumulation leads to local inflammation and insulin resistance (IR). Indeed, overnutrition triggers uncontrolled inflammatory responses in WAT, leading to chronic low-grade inflammation, therefore fostering the progression of IR. This review summarizes the current knowledge on WAT dysfunction in obesity and its associated metabolic abnormalities, such as IR. A better understanding of the mechanisms regulating adipose tissue expansion in obesity is required for the development of future therapeutic approaches in obesity-associated metabolic complications.
Collapse
Affiliation(s)
- Michele Longo
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Federica Zatterale
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Jamal Naderi
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Luca Parrillo
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Gregory Alexander Raciti
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| | - Claudia Miele
- Department of Translational Medicine, Federico II University of Naples, 80131 Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy.
| |
Collapse
|
102
|
Blázquez-Medela AM, Jumabay M, Boström KI. Beyond the bone: Bone morphogenetic protein signaling in adipose tissue. Obes Rev 2019; 20:648-658. [PMID: 30609449 PMCID: PMC6447448 DOI: 10.1111/obr.12822] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/02/2018] [Accepted: 11/25/2018] [Indexed: 02/06/2023]
Abstract
The bone morphogenetic proteins (BMPs) belong to the same superfamily as related to transforming growth factor β (TGFβ), growth and differentiation factors (GDFs), and activins. They were initially described as inducers of bone formation but are now known to be involved in morphogenetic activities and cell differentiation throughout the body, including the development of adipose tissue and adipogenic differentiation. BMP4 and BMP7 are the most studied BMPs in adipose tissue, with major roles in white adipogenesis and brown adipogenesis, respectively, but other BMPs such as BMP2, BMP6, and BMP8b as well as some inhibitors and modulators have been shown to also affect adipogenesis. It has become ever more important to understand adipose regulation, including the BMP pathways, in light of the strong links between obesity and metabolic and cardiovascular disease. In this review, we summarize the available information on BMP signaling in adipose tissue using preferentially articles that have appeared in the last decade, which together demonstrate the importance of BMP signaling in adipose biology.
Collapse
Affiliation(s)
- Ana M Blázquez-Medela
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Medet Jumabay
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States.,Molecular Biology Institute, UCLA, Los Angeles, California, United States
| |
Collapse
|
103
|
Atawia RT, Bunch KL, Toque HA, Caldwell RB, Caldwell RW. Mechanisms of obesity-induced metabolic and vascular dysfunctions. FRONT BIOSCI-LANDMRK 2019; 24:890-934. [PMID: 30844720 PMCID: PMC6689231 DOI: 10.2741/4758] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity has reached epidemic proportions and its prevalence is climbing. Obesity is characterized by hypertrophied adipocytes with a dysregulated adipokine secretion profile, increased recruitment of inflammatory cells, and impaired metabolic homeostasis that eventually results in the development of systemic insulin resistance, a phenotype of type 2 diabetes. Nitric oxide synthase (NOS) is an enzyme that converts L-arginine to nitric oxide (NO), which functions to maintain vascular and adipocyte homeostasis. Arginase is a ureohydrolase enzyme that competes with NOS for L-arginine. Arginase activity/expression is upregulated in obesity, which results in diminished bioavailability of NO, impairing both adipocyte and vascular endothelial cell function. Given the emerging role of NO in the regulation of adipocyte physiology and metabolic capacity, this review explores the interplay between arginase and NO, and their effect on the development of metabolic disorders, cardiovascular diseases, and mitochondrial dysfunction in obesity. A comprehensive understanding of the mechanisms involved in the development of obesity-induced metabolic and vascular dysfunction is necessary for the identification of more effective and tailored therapeutic avenues for their prevention and treatment.
Collapse
Affiliation(s)
- Reem T Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Katharine L Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology,and Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Robert W Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904,USA,
| |
Collapse
|
104
|
Wong JSC, Chu WK, Li BFL, Zhang BN, Pang CP, Chong KKL. Elevated bone morphogenic protein 4 expression implicated in site-specific adipogenesis in thyroid associated orbitopathy. Exp Eye Res 2019; 181:185-189. [PMID: 30721670 DOI: 10.1016/j.exer.2019.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/11/2019] [Accepted: 01/17/2019] [Indexed: 10/27/2022]
Abstract
Periorbital adipose tissue expansion is a key pathological change in thyroid associated orbitopathy (TAO). Bone morphogenic protein 4 (BMP4) is instrumental in adipogenesis. We compared site-specific BMP4 expression and its effect on adipogenesis using donor-matched adipose tissue-derived stromal cells (ADSC) from TAO patients. In this study, ADSC were generated from periorbital (eyelid, orbital) and subcutaneous (abdominal) adipose tissue. BMP4 expression was characterized by RT-PCR and immunofluorescent staining and compared among ADSC from the three anatomic depots. Effects on adipogenesis after knocking down endogenous BMP4 were quantified by adipogenic markers PPARγ and perilipin. Exogenous BMP4 protein was added after BMP4 knockdown to study the role of BMP4 in adipogenesis. Our results showed that BMP4 staining in periorbital adipose tissue was stronger than those in subcutaneous. BMP4 mRNA expression was higher in eyelid (4.4-2489.4-fold) and orbital (6.9-1811-fold) than that of subcutaneous ADSC, whereas expression fell during induced adipogenesis. After BMP4 knockdown, both adipogenic markers PPARγ (eyelid: 1.7-fold, p = 0.038; orbital: 1.4-fold, p = 0.126) and perilipin (eyelid:1.7-fold, p = 0.001; orbital:2.6-fold, p = 0.066) increased in periorbital ADSC upon induction. These increased expression fell after adding exogenous BMP4 protein. Our findings demonstrated higher BMP4 expression was found in periorbital ADSC and adipose tissue compared to donor-matched subcutaneous counterparts, which fell during adipogenic induction. Knocking down BMP4 expression further enhanced adipogenesis in periorbital ADSC. This effect was reversed by adding exogenous BMP4 protein. We suggested a novel role of BMP4 in modulating site-specific adipogenesis in TAO patients.
Collapse
Affiliation(s)
- Janice Siu Chong Wong
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 4/F, Hong Kong Eye Hospital, 147K Argyle Street, Kowloon, Hong Kong, China
| | - Wai Kit Chu
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 4/F, Hong Kong Eye Hospital, 147K Argyle Street, Kowloon, Hong Kong, China
| | - Benjamin Fuk Loi Li
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 4/F, Hong Kong Eye Hospital, 147K Argyle Street, Kowloon, Hong Kong, China
| | - Bi Ning Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 4/F, Hong Kong Eye Hospital, 147K Argyle Street, Kowloon, Hong Kong, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 4/F, Hong Kong Eye Hospital, 147K Argyle Street, Kowloon, Hong Kong, China
| | - Kelvin Kam Lung Chong
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 4/F, Hong Kong Eye Hospital, 147K Argyle Street, Kowloon, Hong Kong, China; Department of Ophthalmology and Visual Sciences, 1/F, Eye Centre, Prince of Wales Hospital, 30-32 Ngan Shing St, Sha Tin, Hong Kong, China.
| |
Collapse
|
105
|
West MD, Chang CF, Larocca D, Li J, Jiang J, Sim P, Labat I, Chapman KB, Wong KE, Nicoll J, Van Kanegan MJ, de Grey ADNJ, Nasonkin IO, Stahl A, Sternberg H. Clonal derivation of white and brown adipocyte progenitor cell lines from human pluripotent stem cells. Stem Cell Res Ther 2019; 10:7. [PMID: 30616682 PMCID: PMC6323697 DOI: 10.1186/s13287-018-1087-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The role of brown fat in non-shivering thermogenesis and the discovery of brown fat depots in adult humans has made it the subject of intense research interest. A renewable source of brown adipocyte (BA) progenitors would be highly valuable for research and therapy. Directed differentiation of human pluripotent stem (hPS) cells to white or brown adipocytes is limited by lack of cell purity and scalability. Here we describe an alternative approach involving the identification of clonal self-renewing human embryonic progenitor (hEP) cell lines following partial hPS cell differentiation and selection of scalable clones. METHODS We screened a diverse panel of hPS cell-derived clonal hEP cell lines for adipocyte markers following growth in adipocyte differentiation medium. The transcriptome of the human hES-derived clonal embryonic progenitor cell lines E3, C4ELS5.1, NP88, and NP110 representing three class of definitive adipocyte progenitors were compared to the relatively non-adipogenic line E85 and adult-derived BAT and SAT-derived cells using gene expression microarrays, RT-qPCR, metabolic analysis and immunocytochemistry. Differentiation conditions were optimized for maximal UCP1 expression. RESULTS Many of the differentiated hEP cell lines expressed the adipocyte marker, FAPB4, but only a small subset expressed definitive adipocyte markers including brown adipocyte marker, UCP1. Class I cells (i.e., E3) expressed CITED1, ADIPOQ, and C19orf80 but little to no UCP1. Class II (i.e., C4ELS5.1) expressed CITED1 and UCP1 but little ADIPOQ and LIPASIN. Class III (i.e., NP88, NP110) expressed CITED1, ADIPOQ, C19orf80, and UCP1 in a similar manner as fetal BAT-derived (fBAT) cells. Differentiated NP88 and NP110 lines were closest to fBAT cells morphologically in adiponectin and uncoupling protein expression. But they were more metabolically active than fBAT cells, had higher levels of 3-hydroxybutyrate, and lacked expression of fetal/adult marker, COX7A1. The hEP BA progenitor lines were scalable to 17 passages without loss of differentiation capacity and could be readily rederived. CONCLUSIONS Taken together, these data demonstrate that self-renewing adipocyte progenitor cells can be derived from hES cells and that they are functionally like BAT cells but with unique properties that might be advantageous for basic research and for development of cell-based treatments for metabolic diseases.
Collapse
Affiliation(s)
- Michael D. West
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Ching-Fang Chang
- 0000 0001 2181 7878grid.47840.3fUniversity of California, Berkeley, CA 94720 USA
| | - Dana Larocca
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Jie Li
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Jianjie Jiang
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Pamela Sim
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Ivan Labat
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| | - Karen B. Chapman
- 0000 0001 2171 9311grid.21107.35Johns Hopkins University, Baltimore, MD 21218 USA
| | - Kari E. Wong
- grid.429438.0Metabolon Inc., Morrisville, NC 27560 USA
| | - James Nicoll
- grid.422945.cZen-Bio, Inc., Research Triangle Park, NC 27709 USA
| | | | - Aubrey D. N. J. de Grey
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA ,SENS Research Foundation, Mountain View, CA 94041 USA
| | | | - Andreas Stahl
- 0000 0001 2181 7878grid.47840.3fUniversity of California, Berkeley, CA 94720 USA
| | - Hal Sternberg
- AgeX Therapeutics, Inc., 1010 Atlantic Ave, Alameda, CA 94501 USA
| |
Collapse
|
106
|
Hammarstedt A, Gogg S, Hedjazifar S, Nerstedt A, Smith U. Impaired Adipogenesis and Dysfunctional Adipose Tissue in Human Hypertrophic Obesity. Physiol Rev 2019; 98:1911-1941. [PMID: 30067159 DOI: 10.1152/physrev.00034.2017] [Citation(s) in RCA: 304] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The subcutaneous adipose tissue (SAT) is the largest and best storage site for excess lipids. However, it has a limited ability to expand by recruiting and/or differentiating available precursor cells. When inadequate, this leads to a hypertrophic expansion of the cells with increased inflammation, insulin resistance, and a dysfunctional prolipolytic tissue. Epi-/genetic factors regulate SAT adipogenesis and genetic predisposition for type 2 diabetes is associated with markers of an impaired SAT adipogenesis and development of hypertrophic obesity also in nonobese individuals. We here review mechanisms for the adipose precursor cells to enter adipogenesis, emphasizing the role of bone morphogenetic protein-4 (BMP-4) and its endogenous antagonist gremlin-1, which is increased in hypertrophic SAT in humans. Gremlin-1 is a secreted and a likely important mechanism for the impaired SAT adipogenesis in hypertrophic obesity. Transiently increasing BMP-4 enhances adipogenic commitment of the precursor cells while maintained BMP-4 signaling during differentiation induces a beige/brown oxidative phenotype in both human and murine adipose cells. Adipose tissue growth and development also requires increased angiogenesis, and BMP-4, as a proangiogenic molecule, may also be an important feedback regulator of this. Hypertrophic obesity is also associated with increased lipolysis. Reduced lipid storage and increased release of FFA by hypertrophic SAT are important mechanisms for the accumulation of ectopic fat in the liver and other places promoting insulin resistance. Taken together, the limited expansion and storage capacity of SAT is a major driver of the obesity-associated metabolic complications.
Collapse
Affiliation(s)
- Ann Hammarstedt
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Silvia Gogg
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Shahram Hedjazifar
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Annika Nerstedt
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Ulf Smith
- Department of Molecular and Clinical Medicine, The Lundberg Laboratory for Diabetes Research, the Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
107
|
Larson CJ. Translational Pharmacology and Physiology of Brown Adipose Tissue in Human Disease and Treatment. Handb Exp Pharmacol 2019; 251:381-424. [PMID: 30689089 DOI: 10.1007/164_2018_184] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Human brown adipose tissue (BAT) is experimentally modeled to better understand the biology of this important metabolic tissue, and also to enable the potential discovery and development of novel therapeutics for obesity and sequelae resulting from the persistent positive energy balance. This chapter focuses on translation into humans of findings and hypotheses generated in nonhuman models of BAT pharmacology. Given the demonstrated challenges of sustainably reducing caloric intake in modern humans, potential solutions to obesity likely lie in increasing energy expenditure. The energy-transforming activities of a single cell in any given tissue can be conceptualized as a flow of chemical energy from energy-rich substrate molecules into energy-expending, endergonic biological work processes through oxidative degradation of organic molecules ingested as nutrients. Despite the relatively tight coupling between metabolic reactions and products, some expended energy is incidentally lost as heat, and in this manner a significant fraction of the energy originally captured from the environment nonproductively transforms into heat rather than into biological work. In human and other mammalian cells, some processes are even completely uncoupled, and therefore purely energy consuming. These molecular and cellular actions sum up at the physiological level to adaptive thermogenesis, the endogenous physiology in which energy is nonproductively released as heat through uncoupling of mitochondria in brown fat and potentially skeletal muscle. Adaptive thermogenesis in mammals occurs in three forms, mostly in skeletal muscle and brown fat: shivering thermogenesis in skeletal muscle, non-shivering thermogenesis in brown fat, and diet-induced thermogenesis in brown fat. At the cellular level, the greatest energy transformations in humans and other eukaryotes occur in the mitochondria, where creating energetic inefficiency by uncoupling the conversion of energy-rich substrate molecules into ATP usable by all three major forms of biological work occurs by two primary means. Basal uncoupling occurs as a passive, general, nonspecific leak down the proton concentration gradient across the membrane in all mitochondria in the human body, a gradient driving a key step in ATP synthesis. Inducible uncoupling, which is the active conduction of protons across gradients through processes catalyzed by proteins, occurs only in select cell types including BAT. Experiments in rodents revealed UCP1 as the primary mammalian molecule accounting for the regulated, inducible uncoupling of BAT, and responsive to both cold and pharmacological stimulation. Cold stimulation of BAT has convincingly translated into humans, and older clinical observations with nonselective 2,4-DNP validate that human BAT's participation in pharmacologically mediated, though nonselective, mitochondrial membrane decoupling can provide increased energy expenditure and corresponding body weight loss. In recent times, however, neither beta-adrenergic antagonism nor unselective sympathomimetic agonism by ephedrine and sibutramine provide convincing evidence that more BAT-selective mechanisms can impact energy balance and subsequently body weight. Although BAT activity correlates with leanness, hypothesis-driven selective β3-adrenergic agonism to activate BAT in humans has only provided robust proof of pharmacologic activation of β-adrenergic receptor signaling, limited proof of the mechanism of increased adaptive thermogenesis, and no convincing evidence that body weight loss through negative energy balance upon BAT activation can be accomplished outside of rodents. None of the five demonstrably β3 selective molecules with sufficient clinical experience to merit review provided significant weight loss in clinical trials (BRL 26830A, TAK 677, L-796568, CL 316,243, and BRL 35135). Broader conclusions regarding the human BAT therapeutic hypothesis are limited by the absence of data from most studies demonstrating specific activation of BAT thermogenesis in most studies. Additionally, more limited data sets with older or less selective β3 agonists also did not provide strong evidence of body weight effects. Encouragingly, β3-adrenergic agonists, catechins, capsinoids, and nutritional extracts, even without robust negative energy balance outcomes, all demonstrated increased total energy expenditure that in some cases could be associated with concomitant activation of BAT, though the absence of body weight loss indicates that in no cases did the magnitude of negative energy balance reach sufficient levels. Glucocorticoid receptor agonists, PPARg agonists, and thyroid hormone receptor agonists all possess defined molecular and cellular pharmacology that preclinical models predicted to be efficacious for negative energy balance and body weight loss, yet their effects on human BAT thermogenesis upon translation were inconsistent with predictions and disappointing. A few new mechanisms are nearing the stage of clinical trials and may yet provide a more quantitatively robust translation from preclinical to human experience with BAT. In conclusion, translation into humans has been demonstrated with BAT molecular pharmacology and cell biology, as well as with physiological response to cold. However, despite pharmacologically mediated, statistically significant elevation in total energy expenditure, translation into biologically meaningful negative energy balance was not achieved, as indicated by the absence of measurable loss of body weight over the duration of a clinical study.
Collapse
Affiliation(s)
- Christopher J Larson
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
108
|
Yu J, Lv Y, Wang F, Kong X, Di W, Liu J, Sheng Y, Lv S, Ding G. MiR-27b-3p Inhibition Enhances Browning of Epididymal Fat in High-Fat Diet Induced Obese Mice. Front Endocrinol (Lausanne) 2019; 10:38. [PMID: 30778336 PMCID: PMC6369196 DOI: 10.3389/fendo.2019.00038] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/17/2019] [Indexed: 11/24/2022] Open
Abstract
Objective: Long-term dysregulation of energy balance is the key component of the obesity epidemic. Given the harm of central obesity and the discovery that beige cells appear within white adipose tissue (WAT), enhancing the energy-expending or "browning" ability of visceral adipose tissue (VAT) has become of therapeutic interest. In this study, we focused on the regulating role of microRNA (miRNA)-27b-3p in mice epididymal white adipose tissue (eWAT) browning. Methods: High-fat diet (HFD) induced obese mice model was constructed. Expression of miR-27b-3p and Ucp1 in eWAT was measured during the course of HFD. Through tail vein injection of antimiR-27b-3p, miR-27b-3p expression was inhibited to analyze the potential role of miR-27b-3p in fat browning and metabolism. Results: miR-27b-3p was predominantly expressed in eWAT and browning ability of eWAT in HFD induced obese mice was impaired. Inhibition of miR-27b-3p enhanced browning capacity of eWAT in mice fed an HFD and led to weight loss and insulin sensitivity improvement. Conclusions: High expression of miR-27b-3p in eWAT inhibits browning ability and leads to visceral fat accumulation. It is suggested miR-27b-3p may become a potential therapeutic option for visceral obesity and its associated diseases.
Collapse
Affiliation(s)
- Jing Yu
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Lv
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fengliang Wang
- Department of Breast Surgery, The Affiliated Obstetrics and Gynaecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiaocen Kong
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjuan Di
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Liu
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunlu Sheng
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shan Lv
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoxian Ding
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Guoxian Ding
| |
Collapse
|
109
|
Hughes MF, Lenighan YM, Godson C, Roche HM. Exploring Coronary Artery Disease GWAs Targets With Functional Links to Immunometabolism. Front Cardiovasc Med 2018; 5:148. [PMID: 30460244 PMCID: PMC6232936 DOI: 10.3389/fcvm.2018.00148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
Finding genetic variants that cause functional disruption or regulatory change among the many implicated GWAs variants remains a key challenge to translating the findings from GWAs to therapeutic treatments. Defining the causal mechanisms behind the variants require functional screening experiments that can be complex and costly. Prioritizing variants for functional characterization using techniques that capture important functional and regulatory elements can assist this. The genetic architecture of complex traits such as cardiovascular disease and type II diabetes comprise an enormously large number of variants of small effect contributing to heritability and spread throughout the genome. This makes it difficult to distinguish which variants or core genes are most relevant for prioritization and how they contribute to the regulatory networks that become dysregulated leading to disease. Despite these challenges, recent GWAs for CAD prioritized genes associated with lipid metabolism, coagulation and adhesion along with novel signals related to innate immunity, adipose tissue and, vascular function as important core drivers of risk. We focus on three examples of novel signals associated with CAD which affect risk through missense or UTR mutations indicating their potential for therapeutic modification. These variants play roles in adipose tissue function vascular function and innate immunity which form the cornerstones of immuno-metabolism. In addition we have explored the putative, but potentially important interactions between the environment, specifically food and nutrition, with respect to key processes.
Collapse
Affiliation(s)
- Maria F Hughes
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,Nutrigenomics Research Group, UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland.,Centre of Excellence for Public Health, Queen's University Belfast, Belfast, United Kingdom.,UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Yvonne M Lenighan
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Helen M Roche
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,Nutrigenomics Research Group, UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland.,UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
110
|
Abstract
During the last decades, research on adipose tissues has spread in parallel with the extension of obesity. Several observations converged on the idea that adipose tissues are organized in a large organ with endocrine and plastic properties. Two parenchymal components: white (WATs) and brown adipose tissues (BATs) are contained in subcutaneous and visceral compartments. Although both have endocrine properties, their function differs: WAT store lipids to allow intervals between meals, BAT burns lipids for thermogenesis. In spite of these opposite functions, they share the ability for reciprocal reversible transdifferentiation to tackle special physiologic needs. Thus, chronic need for thermogenesis induces browning and chronic positive energy balance induce whitening. Lineage tracing and data from explant studies strongly suggest other remodeling properties of this organ. During pregnancy and lactation breast WAT transdifferentiates into milk-secreting glands, composed by cells with abundant cytoplasmic lipids (pink adipocytes) and in the postlactation period pink adipocytes transdifferentiate back into WAT and BAT. The plastic properties of mature adipocytes are supported also by a liposecretion process in vitro where adult cell in culture transdifferentiate to differentiated fibroblast-like elements able to give rise to different phenotypes (rainbow adipocytes). In addition, the inflammasome system is activated in stressed adipocytes from obese adipose tissue. These adipocytes die and debris are reabsorbed by macrophages inducing a chronic low-grade inflammation, potentially contributing to insulin resistance and T2 diabetes. Thus, the plastic properties of this organ could open new therapeutic perspectives in the obesity-related metabolic disease and in breast pathologies. © 2018 American Physiological Society. Compr Physiol 8:1357-1431, 2018.
Collapse
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
111
|
Lee MJ. Transforming growth factor beta superfamily regulation of adipose tissue biology in obesity. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1160-1171. [PMID: 29409985 DOI: 10.1016/j.bbadis.2018.01.025] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/09/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
Accumulation of dysfunctional white adipose tissues increases risks for cardiometabolic diseases in obesity. In addition to white, brown or brite adipose tissues are also present in adult humans and increasing their amount may be protective. Therefore, understanding factors regulating the amount and function of each adipose depot is crucial for developing therapeutic targets for obesity and its associated metabolic diseases. The transforming growth factor beta (TGFβ) superfamily, which consists of TGFβ, BMPs, GDFs, and activins, controls multiple aspects of adipose biology. This review focuses on the recent development in understanding the role of TGFβ superfamily in the regulation of white, brite and brown adipocyte differentiation, adipose tissue fibrosis, and adipocyte metabolic and endocrine functions. TGFβ family and their antagonists are produced locally within adipose tissues and their expression levels are altered in obesity. We also discuss their potential contribution to adipose tissue dysfunction in obesity.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave Levy Place, Box 1152, New York, NY 10029, USA.
| |
Collapse
|
112
|
Wang X, Li L, Zhu C, Gao J, Qu S. Alteration of Bone Mineral Density Differs Between Genders in Obese Subjects After Laparoscopic Sleeve Gastrectomy: Bone Morphogenetic Protein 4 May Count. Obes Surg 2018; 28:3221-3226. [DOI: 10.1007/s11695-018-3298-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
113
|
Hoffmann JM, Grünberg JR, Church C, Elias I, Palsdottir V, Jansson JO, Bosch F, Hammarstedt A, Hedjazifar S, Smith U. BMP4 Gene Therapy in Mature Mice Reduces BAT Activation but Protects from Obesity by Browning Subcutaneous Adipose Tissue. Cell Rep 2018; 20:1038-1049. [PMID: 28768190 DOI: 10.1016/j.celrep.2017.07.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/10/2017] [Accepted: 07/10/2017] [Indexed: 01/02/2023] Open
Abstract
We examined the effect of Bone Morphogenetic Protein 4 (BMP4) on energy expenditure in adult mature mice by targeting the liver with adeno-associated viral (AAV) BMP4 vectors to increase circulating levels. We verified the direct effect of BMP4 in inducing a brown oxidative phenotype in differentiating preadipocytes in vitro. AAV-BMP4-treated mice display marked browning of subcutaneous adipocytes, with increased mitochondria and Uncoupling Protein 1 (UCP1). These mice are protected from obesity on a high-fat diet and have increased whole-body energy expenditure, improved insulin sensitivity, reduced liver fat, and reduced adipose tissue inflammation. On a control diet, they show unchanged body weight but improved insulin sensitivity. In contrast, AAV-BMP4-treated mice showed beiging of BAT with reduced UCP1, increased lipids, and reduced hormone-sensitive lipase (HSL). Thus, BMP4 exerts different effects on WAT and BAT, but the overall effect is to enhance insulin sensitivity and whole-body energy expenditure by browning subcutaneous adipose tissue.
Collapse
Affiliation(s)
- Jenny M Hoffmann
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - John R Grünberg
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 1TN, UK
| | - Christopher Church
- Cardiovascular and Metabolic Disease, MedImmune, Granta Park, Cambridge CB21 6GH, UK
| | - Ivet Elias
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08029 Madrid, Spain
| | - Vilborg Palsdottir
- Department of Physiology/Endocrinology, the Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - John-Olov Jansson
- Department of Physiology/Endocrinology, the Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08029 Madrid, Spain
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Shahram Hedjazifar
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
114
|
Grünberg JR, Elvin J, Paul A, Hedjazifar S, Hammarstedt A, Smith U. CCN5/WISP2 and metabolic diseases. J Cell Commun Signal 2018; 12:309-318. [PMID: 29247377 PMCID: PMC5842198 DOI: 10.1007/s12079-017-0437-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 12/27/2022] Open
Abstract
Obesity and type 2 diabetes increase worldwide at an epidemic rate. It is expected that by the year 2030 around 500 million people will have diabetes; predominantly type 2 diabetes. The CCN family of proteins has become of interest in both metabolic and other common human diseases because of their effects on mesenchymal stem cell (MSCs) proliferation and differentiation as well as being important regulators of fibrosis. We here review current knowledge of the WNT1 inducible signaling pathway protein 2 (CCN5/WISP2). It has been shown to be an important regulator of both these processes through effects on both the canonical WNT and the TGFβ pathways. It is also under normal regulation by the adipogenic commitment factor BMP4, in contrast to conventional canonical WNT ligands, and allows MSCs to undergo normal adipose cell differentiation. CCN5/WISP2 is highly expressed in, and secreted by, MSCs and is an important regulator of MSCs growth. In a transgenic mouse model overexpressing CCN5/WISP2 in the adipose tissue, we have shown that it is secreted and circulating in the blood, the mice develop hypercellular white and brown adipose tissue, have increased lean body mass and enlarged hypercellular hearts. Obese transgenic mice had improved insulin sensitivity. Interestingly, the anti-fibrotic effect of CCN5/WISP2 is protective against heart failure by inhibition of the TGFβ pathway. Understanding how CCN5/WISP2 is regulated and signals is important and may be useful for developing new treatment strategies in obesity and metabolic diseases and it can also be a target in regenerative medicine.
Collapse
Affiliation(s)
- John R Grünberg
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Johannes Elvin
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Alexandra Paul
- Department of Biology and Biological Engineering, Chalmers University of Technology, 41296, Gothenburg, Sweden
| | - Shahram Hedjazifar
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
| |
Collapse
|
115
|
Longo M, Raciti GA, Zatterale F, Parrillo L, Desiderio A, Spinelli R, Hammarstedt A, Hedjazifar S, Hoffmann JM, Nigro C, Mirra P, Fiory F, Formisano P, Miele C, Smith U, Beguinot F. Epigenetic modifications of the Zfp/ZNF423 gene control murine adipogenic commitment and are dysregulated in human hypertrophic obesity. Diabetologia 2018; 61:369-380. [PMID: 29067487 PMCID: PMC6448963 DOI: 10.1007/s00125-017-4471-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/08/2017] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Subcutaneous adipocyte hypertrophy is associated with insulin resistance and increased risk of type 2 diabetes, and predicts its future development independent of obesity. In humans, subcutaneous adipose tissue hypertrophy is a consequence of impaired adipocyte precursor cell recruitment into the adipogenic pathway rather than a lack of precursor cells. The zinc finger transcription factor known as zinc finger protein (ZFP) 423 has been identified as a major determinant of pre-adipocyte commitment and maintained white adipose cell function. Although its levels do not change during adipogenesis, ectopic expression of Zfp423 in non-adipogenic murine cells is sufficient to activate expression of the gene encoding peroxisome proliferator-activated receptor γ (Pparγ; also known as Pparg) and increase the adipogenic potential of these cells. We investigated whether the Zfp423 gene is under epigenetic regulation and whether this plays a role in the restricted adipogenesis associated with hypertrophic obesity. METHODS Murine 3T3-L1 and NIH-3T3 cells were used as fibroblasts committed and uncommitted to the adipocyte lineage, respectively. Human pre-adipocytes were isolated from the stromal vascular fraction of subcutaneous adipose tissue of 20 lean non-diabetic individuals with a wide adipose cell size range. mRNA levels were measured by quantitative real-time PCR, while methylation levels were analysed by bisulphite sequencing. Chromatin structure was analysed by micrococcal nuclease protection assay, and DNA-methyltransferases were chemically inhibited by 5-azacytidine. Adipocyte differentiation rate was evaluated by Oil Red O staining. RESULTS Comparison of uncommitted (NIH-3T3) and committed (3T3-L1) adipose precursor cells revealed that Zfp423 expression increased (p < 0.01) in parallel with the ability of the cells to differentiate into mature adipocytes owing to both decreased promoter DNA methylation (p < 0.001) and nucleosome occupancy (nucleosome [NUC] 1 p < 0.01; NUC2 p < 0.001) in the 3T3-L1 compared with NIH-3T3 cells. Interestingly, non-adipogenic epigenetic profiles can be reverted in NIH-3T3 cells as 5-azacytidine treatment increased Zfp423 mRNA levels (p < 0.01), reduced DNA methylation at a specific CpG site (p < 0.01), decreased nucleosome occupancy (NUC1, NUC2: p < 0.001) and induced adipocyte differentiation (p < 0.05). These epigenetic modifications can also be initiated in response to changes in the pre-adipose cell microenvironment, in which bone morphogenetic protein 4 (BMP4) plays a key role. We finally showed that, in human adipocyte precursor cells, impaired epigenetic regulation of zinc nuclear factor (ZNF)423 (the human orthologue of murine Zfp423) was associated with inappropriate subcutaneous adipose cell hypertrophy. As in NIH-3T3 cells, the normal ZNF423 epigenetic profile was rescued by 5-azacytidine exposure. CONCLUSIONS/INTERPRETATION Our results show that epigenetic events regulate the ability of precursor cells to commit and differentiate into mature adipocytes by modulating ZNF423, and indicate that dysregulation of these mechanisms accompanies subcutaneous adipose tissue hypertrophy in humans.
Collapse
Affiliation(s)
- Michele Longo
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Gregory A Raciti
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Federica Zatterale
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Luca Parrillo
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Antonella Desiderio
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Rosa Spinelli
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Ann Hammarstedt
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Shahram Hedjazifar
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jenny M Hoffmann
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Nigro
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Paola Mirra
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Francesca Fiory
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Pietro Formisano
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Claudia Miele
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francesco Beguinot
- URT Genomics of Diabetes-IEOS, CNR & Department of Translational Medicine, Federico II University of Naples, Via Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
116
|
Li S, Li Y, Xiang L, Dong J, Liu M, Xiang G. Sildenafil induces browning of subcutaneous white adipose tissue in overweight adults. Metabolism 2018; 78:106-117. [PMID: 28986166 DOI: 10.1016/j.metabol.2017.09.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 08/24/2017] [Accepted: 09/19/2017] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To investigate that short-term treatment of sildenafil can induce browning of subcutaneous white adipose tissue (sWAT) in human adults. DESIGN A randomized, double-blinded, placebo-controlled, parallel group trial. METHODS Sixteen eligibility overweight male subjects were recruited, comparing 100mg/day sildenafil versus an identical placebo therapy for 7days. sWAT samples were collected from subjects before and after 7-day sildenafil or placebo interventions. RESULTS The results showed that multilocular UCP1-positive adipocytes existed in sWAT samples from subjects after sildenafil treatment. Compared to before treatment in both group as well as after treatment in placebo, sildenafil significantly decreased adipocyte size, increased the expressions of UCP1 protein and mRNA, mitochondrial density, and leak respiratory capacity in sWAT (p<0.05). Sildenafil also increased plasma cyclic guanosine-3',5'-monophosphate (cGMP) and catecholamine concentrations (p<0.05), and consequently activated the expressions of vasodilator-stimulated phosphoprotein (VASP) and p70 ribosomal S6 kinase 1 (S6K1) (p<0.05). Sildenafil did not activate typical brown fat. CONCLUSIONS The current findings demonstrate that sildenafil can induce browning of sWAT in human, and this action may be through cGMP-dependent protein kinase I and mechanistic/mammalian target of rapamycin (mTOR) signaling pathways. Sldenafil may be a promising treatment for metabolic disease.
Collapse
Affiliation(s)
- Shuguang Li
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Lin Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Jing Dong
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Min Liu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China
| | - Guangda Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuluo Road 627, Wuhan 430070, Hubei Province, China.
| |
Collapse
|
117
|
Abstract
Purpose of Review This review provides a summary of recent insights into the role of the local white adipose tissue (WAT) in systemic sclerosis. Recent Findings Adipocytes located in an interfacial WAT area adjacent to fibrotic lesions have an intermediate phenotype and special properties implicated in fibrotic pathology in systemic sclerosis (SSc). The important role of these cells is recognized in different pathologies, such as wound healing, psoriasis, breast cancer, and prostate cancer. Additionally, both immature and mature adipocytes are involved in the appearance of fibroblast-like cells but exhibit different phenotypes and synthetic properties. Summary Adipocytes from interfacial WAT adjacent to the fibrotic area in SSc are phenotypically different from bulk adipocytes and are involved in pathogenesis of SSc. Immature and mature adipocytes from this WAT layer differentiate into various types of fibroblast-like cells, making the local ratio of immature to mature adipocytes in interfacial WAT of particular importance in SSc pathogenesis.
Collapse
|
118
|
Systems biology reveals uncoupling beyond UCP1 in human white fat-derived beige adipocytes. NPJ Syst Biol Appl 2017; 3:29. [PMID: 28983409 PMCID: PMC5626775 DOI: 10.1038/s41540-017-0027-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/04/2017] [Accepted: 08/23/2017] [Indexed: 01/08/2023] Open
Abstract
Pharmaceutical induction of metabolically active beige adipocytes in the normally energy storing white adipose tissue has potential to reduce obesity. Mitochondrial uncoupling in beige adipocytes, as in brown adipocytes, has been reported to occur via the uncoupling protein 1 (UCP1). However, several previous in vitro characterizations of human beige adipocytes have only measured UCP1 mRNA fold increase, and assumed a direct correlation with metabolic activity. Here, we provide an example of pharmaceutical induction of beige adipocytes, where increased mRNA levels of UCP1 are not translated into increased protein levels, and perform a thorough analysis of this example. We incorporate mRNA and protein levels of UCP1, time-resolved mitochondrial characterizations, and numerous perturbations, and analyze all data with a new fit-for-purpose mathematical model. The systematic analysis challenges the seemingly obvious experimental conclusion, i.e., that UCP1 is not active in the induced cells, and shows that hypothesis testing with iterative modeling and experimental work is needed to sort out the role of UCP1. The analyses demonstrate, for the first time, that the uncoupling capability of human beige adipocytes can be obtained without UCP1 activity. This finding thus opens the door to a new direction in drug discovery that targets obesity and its associated comorbidities. Furthermore, the analysis advances our understanding of how to evaluate UCP1-independent thermogenesis in human beige adipocytes.
Collapse
|
119
|
Gaggini M, Carli F, Gastaldelli A. The color of fat and its central role in the development and progression of metabolic diseases. Horm Mol Biol Clin Investig 2017; 31:hmbci-2017-0060. [PMID: 28942436 DOI: 10.1515/hmbci-2017-0060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/29/2017] [Indexed: 02/06/2023]
Abstract
Excess caloric intake does not always translate to an expansion of the subcutaneous adipose tissue (SAT) and increase in fat mass. It is now recognized that adipocyte type (white, WAT, or brown, BAT), size (large vs. small) and metabolism are important factors for the development of cardiometabolic diseases. When the subcutaneous adipose tissue is not able to expand in response to increased energy intake the excess substrate is stored as visceral adipose tissue or as ectopic fat in tissues as muscle, liver and pancreas. Moreover, adipocytes become dysfunctional (adiposopathy, or sick fat), adipokines secretion is increased, fat accumulates in ectopic sites like muscle and liver and alters insulin signaling, increasing the demand for insulin secretion. Thus, there are some subjects that despite having normal weight have the metabolic characteristics of the obese (NWMO), while some obese expand their SAT and remain metabolically healthy (MHO). In this paper we have reviewed the recent findings that relate the metabolism of adipose tissue and its composition to metabolic diseases. In particular, we have discussed the possible role of dysfunctional adipocytes and adipose tissue resistance to the antilipolytic effect of insulin on the development of impaired glucose metabolism. Finally we have reviewed the possible role of BAT vs. WAT in the alteration of lipid and glucose metabolism and the recent studies that have tried to stimulate browning in human adipose tissue.
Collapse
Affiliation(s)
- Melania Gaggini
- Cardiometabolic Risk Group, Institute of Clinical Physiology - CNR, Pisa, Italy
| | - Fabrizia Carli
- Cardiometabolic Risk Group, Institute of Clinical Physiology - CNR, Pisa, Italy
| | - Amalia Gastaldelli
- Head of Cardiometabolic Risk Group and Mass Spectrometry Laboratory, Institute of Clinical Physiology - CNR, via Moruzzi 1 56100, Pisa, Italy, Phone: +39 050 3152679/80, Fax: +39 050 3152166
| |
Collapse
|
120
|
Abstract
Brown and beige adipocytes arise from distinct developmental origins. Brown adipose tissue (BAT) develops embryonically from precursors that also give to skeletal muscle. Beige fat develops postnatally and is highly inducible. Beige fat recruitment is mediated by multiple mechanisms, including de novo beige adipogenesis and white-to-brown adipocyte transdifferentiaiton. Beige precursors reside around vasculatures, and proliferate and differentiate into beige adipocytes. PDGFRα+Ebf2+ precursors are restricted to beige lineage cells, while another PDGFRα+ subset gives rise to beige adipocytes, white adipocytes, or fibrogenic cells. White adipocytes can be reprogramed and transdifferentiated into beige adipocytes. Brown and beige adipocytes display many similar properties, including multilocular lipid droplets, dense mitochondria, and expression of UCP1. UCP1-mediated thermogenesis is a hallmark of brown/beige adipocytes, albeit UCP1-independent thermogenesis also occurs. Development, maintenance, and activation of BAT/beige fat are guided by genetic and epigenetic programs. Numerous transcriptional factors and coactivators act coordinately to promote BAT/beige fat thermogenesis. Epigenetic reprograming influences expression of brown/beige adipocyte-selective genes. BAT/beige fat is regulated by neuronal, hormonal, and immune mechanisms. Hypothalamic thermal circuits define the temperature setpoint that guides BAT/beige fat activity. Metabolic hormones, paracrine/autocrine factors, and various immune cells also play a critical role in regulating BAT/beige fat functions. BAT and beige fat defend temperature homeostasis, and regulate body weight and glucose and lipid metabolism. Obesity is associated with brown/beige fat deficiency, and reactivation of brown/beige fat provides metabolic health benefits in some patients. Pharmacological activation of BAT/beige fat may hold promise for combating metabolic diseases. © 2017 American Physiological Society. Compr Physiol 7:1281-1306, 2017.
Collapse
Affiliation(s)
- Liangyou Rui
- Department of Molecular and Integrative Physiology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
121
|
|
122
|
Genome wide identification, phylogeny, and expression of bone morphogenetic protein genes in tetraploidized common carp (Cyprinus carpio). Gene 2017. [DOI: 10.1016/j.gene.2017.06.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
123
|
Scheideler M, Herzig S, Georgiadi A. Endocrine and autocrine/paracrine modulators of brown adipose tissue mass and activity as novel therapeutic strategies against obesity and type 2 diabetes. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0043/hmbci-2017-0043.xml. [PMID: 28850545 DOI: 10.1515/hmbci-2017-0043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/28/2017] [Indexed: 12/17/2022]
Abstract
The dramatically increasing world-wide prevalence of obesity is recognized as a risk factor for the development of various diseases. The growing research on the role of adipose tissue in controlling energy homeostasis and insulin sensitivity has revealed that the promotion of brown adipose tissue (BAT) activity and the browning of white adipose tissue (WAT) leads to multiple health benefits and prevents obesity and type 2 diabetes (T2D). Inducible thermogenic adipocytes do exist in adult humans and are linked with increased energy combustion and lower body fat mass. Thus brown adipocytes are currently placed at the center of attention for novel therapeutic strategies against metabolic diseases such as obesity and diabetes. Besides the classical, norepinephrine-mediated sympathetic recruitment and activation of thermogenic adipocytes, a number of novel circulating factors have been recently identified to have a positive or negative impact on thermogenic adipocyte formation and activity. In this review their mechanism of action and the plausible therapeutic applications will be summarized and discussed.
Collapse
|
124
|
Hu J, Christian M. Hormonal factors in the control of the browning of white adipose tissue. Horm Mol Biol Clin Investig 2017; 31:hmbci-2017-0017. [PMID: 28731853 DOI: 10.1515/hmbci-2017-0017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/11/2017] [Indexed: 12/24/2022]
Abstract
Adipose tissue has been historically classified into anabolic white adipose tissue (WAT) and catabolic brown adipose tissue (BAT). Recent studies have revealed the plasticity of WAT, where white adipocytes can be induced into 'brown-like' heat-producing adipocytes (BRITE or beige adipocytes). Recruiting and activating BRITE adipocytes in WAT (so-called 'browning') is believed to provide new avenues for the treatment of obesity-related diseases. A number of hormonal factors have been found to regulate BRITE adipose development and activity through autocrine, paracrine and systemic mechanisms. In this mini-review we will discuss the impact of these factors on the browning process, especially those hormonal factors identified with direct effects on white adipocytes.
Collapse
Affiliation(s)
- Jiamiao Hu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, P.R. China
| | - Mark Christian
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CV4 7AL, Coventry, UK
| |
Collapse
|
125
|
Guénantin AC, Briand N, Capel E, Dumont F, Morichon R, Provost C, Stillitano F, Jeziorowska D, Siffroi JP, Hajjar RJ, Fève B, Hulot JS, Collas P, Capeau J, Vigouroux C. Functional Human Beige Adipocytes From Induced Pluripotent Stem Cells. Diabetes 2017; 66:1470-1478. [PMID: 28270520 PMCID: PMC5440013 DOI: 10.2337/db16-1107] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 03/02/2017] [Indexed: 12/25/2022]
Abstract
Activation of thermogenic beige adipocytes has recently emerged as a promising therapeutic target in obesity and diabetes. Relevant human models for beige adipocyte differentiation are essential to implement such therapeutic strategies. We report a straightforward and efficient protocol to generate functional human beige adipocytes from human induced pluripotent stem cells (hiPSCs). Without overexpression of exogenous adipogenic genes, our method recapitulates an adipogenic developmental pathway through successive mesodermal and adipogenic progenitor stages. hiPSC-derived adipocytes are insulin sensitive and display beige-specific markers and functional properties, including upregulation of thermogenic genes, increased mitochondrial content, and increased oxygen consumption upon activation with cAMP analogs. Engraftment of hiPSC-derived adipocytes in mice produces well-organized and vascularized adipose tissue, capable of β-adrenergic-responsive glucose uptake. Our model of human beige adipocyte development provides a new and scalable tool for disease modeling and therapeutic screening.
Collapse
Affiliation(s)
- Anne-Claire Guénantin
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMR_S938, Centre de Recherche Saint-Antoine, Institute of Cardiometabolism and Nutrition, Paris, France
- Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, U.K
| | - Nolwenn Briand
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMR_S938, Centre de Recherche Saint-Antoine, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Emilie Capel
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMR_S938, Centre de Recherche Saint-Antoine, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Florent Dumont
- Institut Cochin, Université Paris Descartes, INSERM U1016, Paris, France
| | - Romain Morichon
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMR_S938, Centre de Recherche Saint-Antoine, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Claire Provost
- Plateforme LIMP, UMS28 Phénotypage du petit animal, Université Pierre et Marie Curie, Paris, France
| | - Francesca Stillitano
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Dorota Jeziorowska
- Sorbonne Universités, Université Pierre et Marie Curie, UMR_S1166, Institute of Cardiometabolism and Nutrition, France
| | - Jean-Pierre Siffroi
- Sorbonne Universités, Université Pierre et Marie Curie, Assistance Publique-Hôspitaux de Paris, Service de Génétique et d'Embryologie Médicales, Hôpital Trousseau, Paris, France
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Bruno Fève
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMR_S938, Centre de Recherche Saint-Antoine, Institute of Cardiometabolism and Nutrition, Paris, France
- Assistance Publique-Hôspitaux de Paris, Service d'Endocrinologie, Hôpital Saint-Antoine, Paris, France
| | - Jean-Sébastien Hulot
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
- Sorbonne Universités, Université Pierre et Marie Curie, UMR_S1166, Institute of Cardiometabolism and Nutrition, France
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
- Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway
| | - Jacqueline Capeau
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMR_S938, Centre de Recherche Saint-Antoine, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Corinne Vigouroux
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM UMR_S938, Centre de Recherche Saint-Antoine, Institute of Cardiometabolism and Nutrition, Paris, France
- Assistance Publique-Hôspitaux de Paris, Service d'Endocrinologie, Hôpital Saint-Antoine, Paris, France
- Assistance Publique-Hôspitaux de Paris, Laboratoire Commun de Biologie et Génétique Moléculaires, Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
126
|
Montanari T, Pošćić N, Colitti M. Factors involved in white-to-brown adipose tissue conversion and in thermogenesis: a review. Obes Rev 2017; 18:495-513. [PMID: 28187240 DOI: 10.1111/obr.12520] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 12/21/2022]
Abstract
Obesity is the result of energy intake chronically exceeding energy expenditure. Classical treatments against obesity do not provide a satisfactory long-term outcome for the majority of patients. After the demonstration of functional brown adipose tissue in human adults, great effort is being devoted to develop therapies based on the adipose tissue itself, through the conversion of fat-accumulating white adipose tissue into energy-dissipating brown adipose tissue. Anti-obesity treatments that exploit endogenous, pharmacological and nutritional factors to drive such conversion are especially in demand. In the present review, we summarize the current knowledge about the various molecules that can be applied in promoting white-to-brown adipose tissue conversion and energy expenditure and the cellular mechanisms involved.
Collapse
Affiliation(s)
- T Montanari
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - N Pošćić
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - M Colitti
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| |
Collapse
|
127
|
Wang X, Chen J, Li L, Zhu CL, Gao J, Rampersad S, Bu L, Qu S. New association of bone morphogenetic protein 4 concentrations with fat distribution in obesity and Exenatide intervention on it. Lipids Health Dis 2017; 16:70. [PMID: 28376799 PMCID: PMC5379507 DOI: 10.1186/s12944-017-0462-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 03/23/2017] [Indexed: 02/04/2023] Open
Abstract
Background Bone morphogenetic protein 4 (BMP-4) has been proven to regulate white adipogensis. We aimed to demonstrate the correlation of BMP-4 with fat distribution and Exenatide treatment on it. Methods We enrolled 69 obese patients. Anthropometric and metabolic indexes were collected. Fat distribution was measured by dual-energy X-ray absorptiometry. BPM-4 levels were assessed using enzyme-link immunosorbent assay kit. 30 obese patients were treated with Exenatide twice a day. Change in body weight, metabolic-related indices and BPM-4 levels were evaluated after 18 weeks. Results 1) The mean(±SD) BMP-4 levels were 763.98 ± 324.11 pg/ml in the obese. BPM-4 levels were significantly positively correlated with estimated visceral adipose tissue mass in all subjects and also in females (r = 0.377, r = 0.625, respectively,all P < 0.05). BPM-4 levels were also significantly positively correlated with body mass index, hip circumference and total fat% in females (r = 0.375,r = 0.429,r = 0.493,respectively, all P < 0.05). BPM-4 levels were negatively correlated with total cholesterol(TC) in all subjects and males also (r = −0.373,r = −0.332,respectively, all P < 0.05). BPM-4 levels were also significantly positively correlated with free triiodothyronine in males (r = 0.441, P < 0.05). 3) Multivariate analyses showed that TC was risk factor of BMP-4 concentration in males and Est.VAT Area was risk factor of BMP-4 levels in females. 4) BMP-4 levels were significantly higher in the obesity with slightly increased thyroid stimulating hormone(TSH) than the obesity without slightly increased TSH (902.08 ± 354.74 pg/ml vs. 720.24 ± 306.41 pg/ml, P < 0.05). 5) Exenatide treatment leads to a significant decreased in BMP-4 from 860.05 ± 352.65 pg/ml to 649.44 + 277.49 pg/ml independent of weight loss(P < 0.05). Conclusion BMP-4 levels were associated with the visceral adipose tissue and may play a certain role in fat distribution and subclinical hypothyroidism in obesity. Exenatide treatment reduced BMP-4 levels independent of weight loss. Trial registration Clinicaltrials.gov Identifier: NCT02118376, Registered 16 April.
Collapse
Affiliation(s)
- Xingchun Wang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiaqi Chen
- Department of Endocrinology and Metabolism, Suzhou Municipal Hospital, Suzhou, Jiangsu Province, 215000, China
| | - Liang Li
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Cui Ling Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jingyang Gao
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Sharvan Rampersad
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Le Bu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
128
|
Davies OG, Liu Y, Player DJ, Martin NRW, Grover LM, Lewis MP. Defining the Balance between Regeneration and Pathological Ossification in Skeletal Muscle Following Traumatic Injury. Front Physiol 2017; 8:194. [PMID: 28421001 PMCID: PMC5376571 DOI: 10.3389/fphys.2017.00194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/15/2017] [Indexed: 12/15/2022] Open
Abstract
Heterotopic ossification (HO) is characterized by the formation of bone at atypical sites. This type of ectopic bone formation is most prominent in skeletal muscle, most frequently resulting as a consequence of physical trauma and associated with aberrant tissue regeneration. The condition is debilitating, reducing a patient's range of motion and potentially causing severe pathologies resulting from nerve and vascular compression. Despite efforts to understand the pathological processes governing HO, there remains a lack of consensus regarding the micro-environmental conditions conducive to its formation, and attempting to define the balance between muscle regeneration and pathological ossification remains complex. The development of HO is thought to be related to a complex interplay between factors released both locally and systemically in response to trauma. It develops as skeletal muscle undergoes significant repair and regeneration, and is likely to result from the misdirected differentiation of endogenous or systemically derived progenitors in response to biochemical and/or environmental cues. The process can be sequentially delineated by the presence of inflammation, tissue breakdown, adipogenesis, hypoxia, neo-vasculogenesis, chondrogenesis and ossification. However, exactly how each of these stages contributes to the formation of HO is at present not well understood. Our previous review examined the cellular contribution to HO. Therefore, the principal aim of this review will be to comprehensively outline changes in the local tissue micro-environment following trauma, and identify how these changes can alter the balance between skeletal muscle regeneration and ectopic ossification. An understanding of the mechanisms governing this condition is required for the development and advancement of HO prophylaxis and treatment, and may even hold the key to unlocking novel methods for engineering hard tissues.
Collapse
Affiliation(s)
- Owen G Davies
- School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK.,School of Chemical Engineering, University of BirminghamBirmingham, UK
| | - Yang Liu
- Wolfson School of Mechanical and Manufacturing Engineering, Loughborough UniversityLoughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK
| | - Liam M Grover
- School of Chemical Engineering, University of BirminghamBirmingham, UK
| | - Mark P Lewis
- National Centre for Sport and Exercise Medicine, Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, School of Sport, Exercise and Health Sciences, Loughborough UniversityLoughborough, UK
| |
Collapse
|
129
|
Modica S, Wolfrum C. The dual role of BMP4 in adipogenesis and metabolism. Adipocyte 2017; 6:141-146. [PMID: 28425843 PMCID: PMC5477726 DOI: 10.1080/21623945.2017.1287637] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/12/2017] [Accepted: 01/22/2017] [Indexed: 12/13/2022] Open
Abstract
BMP4 has a well-established role in triggering commitment of mesenchymal stem cells into the osteogenic and adipogenic linage. We recently described an additional dual function in adipogenesis: after promoting the formation of both white and brown pre-adipocytes, Bmp4 drives terminal differentiation into mature white rather than brown fat cells. Besides this, Bmp4 seems to have a dual role in metabolism either promoting or repressing oxidative metabolism in a cell context dependent manner.
Collapse
Affiliation(s)
- Salvatore Modica
- a Swiss Federal Institute of Technology, Department of Health Science , Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology , Schwerzenbach , Switzerland
| | - Christian Wolfrum
- a Swiss Federal Institute of Technology, Department of Health Science , Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology , Schwerzenbach , Switzerland
| |
Collapse
|
130
|
Overexpressing the novel autocrine/endocrine adipokine WISP2 induces hyperplasia of the heart, white and brown adipose tissues and prevents insulin resistance. Sci Rep 2017; 7:43515. [PMID: 28240264 PMCID: PMC5327486 DOI: 10.1038/srep43515] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/27/2017] [Indexed: 12/31/2022] Open
Abstract
WISP2 is a novel adipokine, most highly expressed in the adipose tissue and primarily in undifferentiated mesenchymal cells. As a secreted protein, it is an autocrine/paracrine activator of canonical WNT signaling and, as an intracellular protein, it helps to maintain precursor cells undifferentiated. To examine effects of increased WISP2 in vivo, we generated an aP2-WISP2 transgenic (Tg) mouse. These mice had increased serum levels of WISP2, increased lean body mass and whole body energy expenditure, hyperplastic brown/white adipose tissues and larger hyperplastic hearts. Obese Tg mice remained insulin sensitive, had increased glucose uptake by adipose cells and skeletal muscle in vivo and ex vivo, increased GLUT4, increased ChREBP and markers of adipose tissue lipogenesis. Serum levels of the novel fatty acid esters of hydroxy fatty acids (FAHFAs) were increased and transplantation of Tg adipose tissue improved glucose tolerance in recipient mice supporting a role of secreted FAHFAs. The growth-promoting effect of WISP2 was shown by increased BrdU incorporation in vivo and Tg serum increased mesenchymal precursor cell proliferation in vitro. In contrast to conventional canonical WNT ligands, WISP2 expression was inhibited by BMP4 thereby allowing normal induction of adipogenesis. WISP2 is a novel secreted regulator of mesenchymal tissue cellularity.
Collapse
|
131
|
Zhang T, Zhang X, Han K, Zhang G, Wang J, Xie K, Xue Q, Fan X. Analysis of long noncoding RNA and mRNA using RNA sequencing during the differentiation of intramuscular preadipocytes in chicken. PLoS One 2017; 12:e0172389. [PMID: 28199418 PMCID: PMC5310915 DOI: 10.1371/journal.pone.0172389] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/03/2017] [Indexed: 02/04/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) regulate metabolic tissue development and function, including adipogenesis. However, little is known about the function and profile of lncRNAs in intramuscular preadipocyte differentiation in chicken. Here, we identified lncRNAs in chicken intramuscular preadipocytes at different differentiation stages using RNA sequencing. A total of 1,311,382,604 clean reads and 25,435 lncRNAs were obtained from 12 samples. In total, 7,433 differentially expressed genes (4,698 lncRNAs and 2,735 mRNAs) were identified by pairwise comparison. These 7,433 differentially expressed genes were grouped into 11 clusters based on their expression patterns by K-means clustering. Using Weighted Gene Coexpression Network Analysis, we identified four stage-specific modules positively related to I0, I2, I4, and I6 stages and two stage-specific modules negatively related to I0 and I2 stages, respectively. Many well-known and novel pathways associated with intramuscular preadipocyte differentiation were identified. We also identified hub genes in each stage-specific module and visualized them in Cytoscape. Our analysis revealed many highly-connected genes, including XLOC_058593, BMP3, MYOD1, and LAMP3. This study provides a valuable resource for chicken lncRNA study and improves our understanding of the biology of preadipocyte differentiation in chicken.
Collapse
Affiliation(s)
- Tao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Xiangqian Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Kunpeng Han
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Genxi Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Jinyu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, Jiangsu, China
- * E-mail:
| | - Kaizhou Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Qian Xue
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Xiaomei Fan
- Vazyme Biotech Co.,Ltd., Economic and Technological Development Zone, Nanjing, Jiangsu, China
| |
Collapse
|
132
|
Laminin differentially regulates the stemness of type I and type II pericytes. Stem Cell Res Ther 2017; 8:28. [PMID: 28173861 PMCID: PMC5297126 DOI: 10.1186/s13287-017-0479-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 01/22/2023] Open
Abstract
Background Laminin, a major basement membrane component that has direct contact with pericytes under physiological conditions, actively regulates the proliferation and differentiation/fate determination of pericytes. Recently, two types of pericytes (type I and type II) with different molecular markers and functions have been identified in skeletal muscles. Whether laminin differentially regulates the proliferation and differentiation of these two subpopulations remains unclear. Methods Wild-type and pericytic laminin-deficient mice under Nestin-GFP background were used to determine if laminin differentially regulates the proliferation and differentiation of type I and type II pericytes. Specifically, type I and type II pericytes were isolated from these mice, and their proliferation and differentiation were examined in vitro. Moreover, in vivo studies were also performed. Results We demonstrate that, although laminin inhibits the proliferation of both type I and type II pericytes in vitro, loss of laminin predominantly induces proliferation of type II pericytes in vivo. In addition, laminin negatively regulates the adipogenic differentiation of type I pericytes and positively regulates the myogenic differentiation of type II pericytes in vitro. Conclusions Laminin differentially regulates the proliferation and differentiation of type I and type II pericytes. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0479-4) contains supplementary material, which is available to authorized users.
Collapse
|
133
|
Zhang Y, Yu M, Dai M, Chen C, Tang Q, Jing W, Wang H, Tian W. miR-450a-5p within rat adipose tissue exosome-like vesicles promotes adipogenic differentiation by targeting WISP2. J Cell Sci 2017; 130:1158-1168. [PMID: 28167681 DOI: 10.1242/jcs.197764] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/31/2017] [Indexed: 02/05/2023] Open
Abstract
Adipose tissue is an active endocrine organ that can secrete a wide number of factors to regulate adipogenesis via paracrine signals. In addition to soluble proteins in adipose tissue, microRNAs (miRNAs) enriched in extracellular vesicles (EVs), such as exosomes or microvesicles, could modulate intercellular communications. In this study, we demonstrated that exosome-like vesicles derived from adipose tissue (Exo-AT) were internalized by adipose tissue-derived stem cells (ADSCs), and that these, in turn, induced adipogenesis. High-throughput sequencing showed that 45 miRNAs were enriched in Exo-AT, and 31.11% of them were associated with adipogenesis, compared with ADSC-derived exosome-like vesicles (Exo-ADSC). miR-450a-5p, one of the most abundant miRNAs in Exo-AT, was a proadipogenic miRNA. Further study demonstrated that miR-450a-5p promoted adipogenesis through repressing expression of WISP2 by targeting its 3' untranslated region. Additionally, Exo-AT could also downregulate the expression of WISP2, while miR-450a-5p inhibitor reversed this effect. Moreover, inhibition of miR-450a-5p impaired adipogenesis mediated by exosome-like vesicles. In conclusion, Exo-AT mediates adipogenic differentiation through a mechanism involving transfer of miR-450a-5p.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mei Yu
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China
| | - Minjia Dai
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chang Chen
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qi Tang
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wei Jing
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hang Wang
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu 610041, China .,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, 3rd Sec., Ren Min Nan Road, Chengdu, Sichuan Province 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
134
|
Rodríguez A, Becerril S, Ezquerro S, Méndez-Giménez L, Frühbeck G. Crosstalk between adipokines and myokines in fat browning. Acta Physiol (Oxf) 2017; 219:362-381. [PMID: 27040995 DOI: 10.1111/apha.12686] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/21/2016] [Accepted: 04/01/2016] [Indexed: 12/20/2022]
Abstract
Skeletal muscle is the largest organ determining whole-body insulin sensitivity and metabolic homoeostasis. Adaptive changes of skeletal muscle in response to physical activity include adjustments in the production and secretion of muscle-derived bioactive factors, known as myokines, such as myostatin, IL-4, IL-6, IL-7 and IL-15, myonectin, follistatin-like 1 or leukaemia inhibitory factor. These myokines not only act locally in the muscle in an autocrine/paracrine manner, but also are released to the bloodstream as endocrine factors to regulate physiological processes in other tissues. Irisin, derived from the cleavage of FNDC5 protein, constitutes a myokine that induces myogenesis and fat browning (switch of white adipocytes to brown fat-like cells) together with a concomitant increase in energy expenditure. Besides being a target for irisin actions, the adipose tissue also constitutes a production site of FNDC5. Interestingly, irisin secretion from subcutaneous and visceral fat depots is decreased by long-term exercise training and fasting, suggesting a discordant regulation of FNDC5/irisin in skeletal muscle and adipose tissue. Accordingly, our group has recently reported that the adipokine leptin differentially regulates FNDC5/irisin expression in skeletal muscle and fat, confirming the crosstalk between both tissues. Moreover, irisin secretion and function are regulated by other myokines, such as follistatin or myostatin, as well as by other adipokines, including fibroblast growth factor 21 and leptin. Taken together, myokines have emerged as novel molecular mediators of fat browning and their activity can be modulated by adipokines, confirming the crosstalk between skeletal muscle and adipose tissue to regulate thermogenesis and energy expenditure.
Collapse
Affiliation(s)
- A. Rodríguez
- Metabolic Research Laboratory; Clínica Universidad de Navarra; Pamplona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
- Obesity & Adipobiology Group; Instituto de Investigación Sanitaria de Navarra (IdiSNA); Pamplona Spain
| | - S. Becerril
- Metabolic Research Laboratory; Clínica Universidad de Navarra; Pamplona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
- Obesity & Adipobiology Group; Instituto de Investigación Sanitaria de Navarra (IdiSNA); Pamplona Spain
| | - S. Ezquerro
- Metabolic Research Laboratory; Clínica Universidad de Navarra; Pamplona Spain
| | - L. Méndez-Giménez
- Metabolic Research Laboratory; Clínica Universidad de Navarra; Pamplona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
- Obesity & Adipobiology Group; Instituto de Investigación Sanitaria de Navarra (IdiSNA); Pamplona Spain
| | - G. Frühbeck
- Metabolic Research Laboratory; Clínica Universidad de Navarra; Pamplona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
- Obesity & Adipobiology Group; Instituto de Investigación Sanitaria de Navarra (IdiSNA); Pamplona Spain
- Department of Endocrinology & Nutrition; Clínica Universidad de Navarra; Pamplona Spain
| |
Collapse
|
135
|
Abstract
Brown adipose tissue (BAT) is the main site of adaptive thermogenesis and experimental studies have associated BAT activity with protection against obesity and metabolic diseases, such as type 2 diabetes mellitus and dyslipidaemia. Active BAT is present in adult humans and its activity is impaired in patients with obesity. The ability of BAT to protect against chronic metabolic disease has traditionally been attributed to its capacity to utilize glucose and lipids for thermogenesis. However, BAT might also have a secretory role, which could contribute to the systemic consequences of BAT activity. Several BAT-derived molecules that act in a paracrine or autocrine manner have been identified. Most of these factors promote hypertrophy and hyperplasia of BAT, vascularization, innervation and blood flow, processes that are all associated with BAT recruitment when thermogenic activity is enhanced. Additionally, BAT can release regulatory molecules that act on other tissues and organs. This secretory capacity of BAT is thought to be involved in the beneficial effects of BAT transplantation in rodents. Fibroblast growth factor 21, IL-6 and neuregulin 4 are among the first BAT-derived endocrine factors to be identified. In this Review, we discuss the current understanding of the regulatory molecules (the so-called brown adipokines or batokines) that are released by BAT that influence systemic metabolism and convey the beneficial metabolic effects of BAT activation. The identification of such adipokines might also direct drug discovery approaches for managing obesity and its associated chronic metabolic diseases.
Collapse
Affiliation(s)
- Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Rubén Cereijo
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Joan Villarroya
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| |
Collapse
|
136
|
Smith U, Kahn BB. Adipose tissue regulates insulin sensitivity: role of adipogenesis, de novo lipogenesis and novel lipids. J Intern Med 2016; 280:465-475. [PMID: 27699898 PMCID: PMC5218584 DOI: 10.1111/joim.12540] [Citation(s) in RCA: 358] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity, the major cause of the current global epidemic of type 2 diabetes (T2D), induces insulin resistance in peripheral insulin target tissues. Several mechanisms have been identified related to cross-talk between adipose tissue, skeletal muscle and liver. These mechanisms involve both increased free fatty acid release and altered secretion of adipokines from adipose tissue. A major determinant of metabolic health is the ability of subcutaneous adipose tissue (SAT) to store excess fat rather than allowing it to accumulate in ectopic depots including liver (i.e. in nonalcoholic fatty liver disease), muscle and heart, or in epicardial/pericardial and visceral fat depots which promote the metabolic complications of obesity. The ability to recruit and differentiate precursor cells into adipose cells (adipogenesis) in SAT is under genetic regulation and is reduced in high-risk individuals who have first-degree relatives with T2D. Early recruitment of new adipose cells is dependent on the cross-talk between canonical WNT and BMP4 signalling; WNT enhances their undifferentiated and proliferative state whereas BMP4 induces their commitment to the adipogenic lineage. Dysregulation of these signalling pathways is associated with impaired adipogenesis and impaired ability to respond to the need to store excess lipids in SAT. This leads to hypertrophic, dysfunctional and insulin-resistant adipose cells with a reduced content of GLUT4, the major insulin-regulated glucose transporter, which in turn reduces adipose tissue glucose uptake and de novo lipogenesis. We recently identified that reduced GLUT4 and lipogenesis in adipocytes impairs the synthesis of a novel family of lipids secreted by adipose tissue (and potentially other tissues), branched fatty acid esters of hydroxy fatty acids (FAHFAs). FAHFAs have beneficial metabolic effects, including enhancing insulin-stimulated glucose transport and glucose-stimulated GLP1 and insulin secretion, as well as powerful anti-inflammatory effects. FAHFA levels are reduced in subcutaneous adipose tissue in insulin-resistant individuals, and this novel family of lipids may become of future therapeutic use.
Collapse
Affiliation(s)
- U Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - B B Kahn
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
137
|
Ponnusamy S, Tran QT, Harvey I, Smallwood HS, Thiyagarajan T, Banerjee S, Johnson DL, Dalton JT, Sullivan RD, Miller DD, Bridges D, Narayanan R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue. FASEB J 2016; 31:266-281. [PMID: 27733447 DOI: 10.1096/fj.201600787rr] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/22/2016] [Indexed: 01/03/2023]
Abstract
Most satiety-inducing obesity therapeutics, despite modest efficacy, have safety concerns that underscore the need for effective peripherally acting drugs. An attractive therapeutic approach for obesity is to optimize/maximize energy expenditure by increasing energy-utilizing thermogenic brown adipose tissue. We used in vivo and in vitro models to determine the role of estrogen receptor β (ER-β) and its ligands on adipose biology. RNA sequencing and metabolomics were used to determine the mechanism of action of ER-β and its ligands. Estrogen receptor β (ER-β) and its selective ligand reprogrammed preadipocytes and precursor stem cells into brown adipose tissue and increased mitochondrial respiration. An ER-β-selective ligand increased markers of tricarboxylic acid-dependent and -independent energy biogenesis and oxygen consumption in mice without a concomitant increase in physical activity or food consumption, all culminating in significantly reduced weight gain and adiposity. The antiobesity effects of ER-β ligand were not observed in ER-β-knockout mice. Serum metabolite profiles of adult lean and juvenile mice were comparable, while that of adult obese mice was distinct, indicating a possible impact of obesity on age-dependent metabolism. This phenotype was partially reversed by ER-β-selective ligand. These data highlight a new role for ER-β in adipose biology and its potential to be a safer alternative peripheral therapeutic target for obesity.-Ponnusamy, S., Tran, Q. T., Harvey, I., Smallwood, H. S., Thiyagarajan, T., Banerjee, S., Johnson, D. L., Dalton, J. T., Sullivan, R. D., Miller, D. D., Bridges, D., Narayanan, R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Quynh T Tran
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Innocence Harvey
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thirumagal Thiyagarajan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Daniel L Johnson
- Molecular Informatics Core, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James T Dalton
- Preclinical Research and Development, GTx, Incorporated, Memphis, Tennessee, USA
| | - Ryan D Sullivan
- Department of Comparative Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; and
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dave Bridges
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; .,West Cancer Center, Memphis, Tennessee, USA
| |
Collapse
|
138
|
Guiu-Jurado E, Unthan M, Böhler N, Kern M, Landgraf K, Dietrich A, Schleinitz D, Ruschke K, Klöting N, Faßhauer M, Tönjes A, Stumvoll M, Körner A, Kovacs P, Blüher M. Bone morphogenetic protein 2 (BMP2) may contribute to partition of energy storage into visceral and subcutaneous fat depots. Obesity (Silver Spring) 2016; 24:2092-100. [PMID: 27515773 DOI: 10.1002/oby.21571] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/21/2016] [Accepted: 05/03/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Bone morphogenetic proteins (BMPs) are important regulators of adipogenesis and may play a role in obesity. In this study, the hypothesis that BMP2 is related to adipose tissue (AT) distribution in obesity was tested. METHODS BMP2 serum concentration (n = 439) and BMP2 and Schnurri-1 and -2 mRNA expression were measured in paired samples of visceral and subcutaneous AT from 547 individuals with a wide range of body mass index. In addition, a single nucleotide polymorphism rs979012 in the BMP2 gene was genotyped for subsequent association studies on quantitative traits related to obesity in 631 individuals. RESULTS BMP2 and Schnurri-1 mRNA were significantly higher in visceral compared with subcutaneous AT. Compared with individuals who were healthy and lean, BMP2 expression in both depots was significantly higher in people with obesity. Significantly higher BMP2 serum concentrations were found in patients with type 2 diabetes with moderate but not morbid obesity. Schnurri-1 and -2 mRNA expression was not related to either BMP2 expression or circulating BMP2. Finally, rs979012 showed nominal association with body mass index and total cholesterol levels. CONCLUSIONS Data suggest that with increasing demand to store excessive energy, AT BMP2 expression increases and may contribute to partitioning of energy storage into visceral and subcutaneous AT depots.
Collapse
Affiliation(s)
- Esther Guiu-Jurado
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Departament de Medicina i Cirurgia, Grup de recerca GEMMAIR (AGAUR)-Medicina Aplicada, Universitat Rovira i Virgili (URV), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Mark Unthan
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Nina Böhler
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Matthias Kern
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Kathrin Landgraf
- Department of Pediatric Endocrinology, Center for Pediatric Research Leipzig (CPL), Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
| | - Arne Dietrich
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Dorit Schleinitz
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Karen Ruschke
- AG Knaus, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Nora Klöting
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | | | - Anke Tönjes
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Pediatric Endocrinology, Center for Pediatric Research Leipzig (CPL), Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Core Unit, Animal Models of Obesity, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany.
- Department of Medicine, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
139
|
Roy B, Curtis ME, Fears LS, Nahashon SN, Fentress HM. Molecular Mechanisms of Obesity-Induced Osteoporosis and Muscle Atrophy. Front Physiol 2016; 7:439. [PMID: 27746742 PMCID: PMC5040721 DOI: 10.3389/fphys.2016.00439] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 09/15/2016] [Indexed: 12/19/2022] Open
Abstract
Obesity and osteoporosis are two alarming health disorders prominent among middle and old age populations, and the numbers of those affected by these two disorders are increasing. It is estimated that more than 600 million adults are obese and over 200 million people have osteoporosis worldwide. Interestingly, both of these abnormalities share some common features including a genetic predisposition, and a common origin: bone marrow mesenchymal stromal cells. Obesity is characterized by the expression of leptin, adiponectin, interleukin 6 (IL-6), interleukin 10 (IL-10), monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), macrophage colony stimulating factor (M-CSF), growth hormone (GH), parathyroid hormone (PTH), angiotensin II (Ang II), 5-hydroxy-tryptamine (5-HT), Advance glycation end products (AGE), and myostatin, which exert their effects by modulating the signaling pathways within bone and muscle. Chemical messengers (e.g., TNF-α, IL-6, AGE, leptins) that are upregulated or downregulated as a result of obesity have been shown to act as negative regulators of osteoblasts, osteocytes and muscles, as well as positive regulators of osteoclasts. These additive effects of obesity ultimately increase the risk for osteoporosis and muscle atrophy. The aim of this review is to identify the potential cellular mechanisms through which obesity may facilitate osteoporosis, muscle atrophy and bone fractures.
Collapse
Affiliation(s)
- Bipradas Roy
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Mary E Curtis
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Letimicia S Fears
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Samuel N Nahashon
- Department of Agricultural and Environmental Sciences, Tennessee State University Nashville, TN, USA
| | - Hugh M Fentress
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| |
Collapse
|
140
|
Wang CL, Xiao F, Wang CD, Zhu JF, Shen C, Zuo B, Wang H, Li D, Wang XY, Feng WJ, Li ZK, Hu GL, Zhang X, Chen XD. Gremlin2 Suppression Increases the BMP-2-Induced Osteogenesis of Human Bone Marrow-Derived Mesenchymal Stem Cells Via the BMP-2/Smad/Runx2 Signaling Pathway. J Cell Biochem 2016; 118:286-297. [PMID: 27335248 DOI: 10.1002/jcb.25635] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/22/2016] [Indexed: 12/12/2022]
Abstract
Osteoblasts are essential for maintaining skeletal architecture and modulating bone microenvironment homeostasis. From numerous associated investigations, the BMP-2 pathway has been well-defined as a vital positive modulator of bone homeostasis. Gremlin2 (Grem2) is a bone morphogenetic protein (BMP) antagonists. However, the effect of Grem2 on the BMP-2-induced osteogenesis of human bone marrow-derived mesenchymal stem cells (hBMSCs) remains ambiguous. This study aimed to analyze the procedure in vitro and in vivo. The differentiation of hBMSCs was assessed by determining the expression levels of several osteoblastic genes, as well as the enzymatic activity and calcification of alkaline phosphatase. We found that Grem2 expression was upregulated by BMP-2 within the range of 0-1 μg/mL, and significant increases were evident at 48, 72, and 96 h after BMP-2 treatment. Si-Grem2 increased the BMP-2-induced osteogenic differentiation of hBMSCs, whereas overexpression of Grem2 had the opposite trend. The result was confirmed using a defective femur model. We also discovered that the BMP-2/Smad/Runx2 pathway played an important role in the process. This study showed that si-Grem2 increased the BMP-2-induced osteogenic differentiation of hBMSCs via the BMP-2/Smad/Runx2 pathway. J. Cell. Biochem. 118: 286-297, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cheng-Long Wang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Fei Xiao
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Chuan-Dong Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences, Shanghai, China
| | - Jun-Feng Zhu
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Chao Shen
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Bin Zuo
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Hui Wang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - De Li
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Xu-Yi Wang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Wei-Jia Feng
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Zhuo-Kai Li
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Guo-Li Hu
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences, Shanghai, China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.,Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Dong Chen
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| |
Collapse
|
141
|
Lv Y, Zhang SY, Liang X, Zhang H, Xu Z, Liu B, Xu MJ, Jiang C, Shang J, Wang X. Adrenomedullin 2 Enhances Beiging in White Adipose Tissue Directly in an Adipocyte-autonomous Manner and Indirectly through Activation of M2 Macrophages. J Biol Chem 2016; 291:23390-23402. [PMID: 27621315 DOI: 10.1074/jbc.m116.735563] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Indexed: 12/15/2022] Open
Abstract
Adrenomedullin 2 (ADM2) is an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family. Our previous studies showed that overexpression of ADM2 in mice reduced obesity and insulin resistance by increasing thermogenesis in brown adipose tissue. However, the effects of ADM2 in another type of thermogenic adipocyte, beige adipocytes, remain to be understood. The plasma ADM2 levels were inversely correlated with obesity in humans, and adipo-ADM2-transgenic (tg) mice displayed resistance to high-fat diet-induced obesity with increased energy expenditure. Beiging of subcutaneous white adipose tissues (WAT) was more noticeably induced in high-fat diet-fed transgenic mice with adipocyte-ADM2 overexpression (adipo-ADM2-tg mice) than in WT animals. ADM2 treatment in primary rat subcutaneous adipocytes induced beiging with up-regulation of UCP1 and beiging-related marker genes and increased mitochondrial uncoupling respiration, which was mainly mediated by activation of the calcitonin receptor-like receptor (CRLR)·receptor activity-modifying protein 1 (RAMP1) complex and PKA and p38 MAPK signaling pathways. Importantly, this adipocyte-autonomous beiging effect by ADM2 was translatable to human primary adipocytes. In addition, M2 macrophage activation also contributed to the beiging effects of ADM2 through catecholamine secretion. Therefore, our study reveals that ADM2 enhances subcutaneous WAT beiging via a direct effect by activating the CRLR·RAMP1-cAMP/PKA and p38 MAPK pathways in white adipocytes and via an indirect effect by stimulating alternative M2 polarization in macrophages. Through both mechanisms, beiging of WAT by ADM2 results in increased energy expenditure and reduced obesity, suggesting ADM2 as a novel anti-obesity target.
Collapse
Affiliation(s)
- Ying Lv
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Song-Yang Zhang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Xianyi Liang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Heng Zhang
- the Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhi Xu
- the Department of General Surgery, Peking University Third Hospital, Beijing 100191, China, and
| | - Bo Liu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Ming-Jiang Xu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Changtao Jiang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China,
| | - Jin Shang
- the Department of Cardiometabolic Disease, Merck Research Laboratories, Merck & Co, Inc., Kenilworth, New Jersey 07033
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|
142
|
Adami GF, Scopinaro N, Cordera R. Adipokine Pattern After Bariatric Surgery: Beyond the Weight Loss. Obes Surg 2016; 26:2793-2801. [DOI: 10.1007/s11695-016-2347-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
143
|
Modica S, Straub LG, Balaz M, Sun W, Varga L, Stefanicka P, Profant M, Simon E, Neubauer H, Ukropcova B, Ukropec J, Wolfrum C. Bmp4 Promotes a Brown to White-like Adipocyte Shift. Cell Rep 2016; 16:2243-2258. [PMID: 27524617 DOI: 10.1016/j.celrep.2016.07.048] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/25/2016] [Accepted: 07/19/2016] [Indexed: 11/19/2022] Open
Abstract
While Bmp4 has a well-established role in the commitment of mesenchymal stem cells into the adipogenic lineage, its role in brown adipocyte formation and activity is not well defined. Here, we show that Bmp4 has a dual function in adipogenesis by inducing adipocyte commitment while inhibiting the acquisition of a brown phenotype during terminal differentiation. Selective brown adipose tissue overexpression of Bmp4 in mice induces a shift from a brown to a white-like adipocyte phenotype. This effect is mediated by Smad signaling and might be in part due to suppression of lipolysis, via regulation of hormone sensitive lipase expression linked to reduced Ppar activity. Given that we observed a strong correlation between BMP4 levels and adipocyte size, as well as insulin sensitivity in humans, we propose that Bmp4 is an important factor in the context of obesity and type 2 diabetes.
Collapse
MESH Headings
- Adipocytes, Brown/cytology
- Adipocytes, Brown/drug effects
- Adipocytes, Brown/metabolism
- Adipocytes, White/cytology
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Adipogenesis/drug effects
- Adipogenesis/genetics
- Adipose Tissue, Brown/cytology
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Animals
- Bone Morphogenetic Protein 4/genetics
- Bone Morphogenetic Protein 4/metabolism
- Bone Morphogenetic Protein 4/pharmacology
- Cell Differentiation
- Cell Line, Transformed
- Cyclic AMP/pharmacology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Regulation
- Humans
- Insulin Resistance
- Male
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, Inbred C57BL
- Peroxisome Proliferator-Activated Receptors/genetics
- Peroxisome Proliferator-Activated Receptors/metabolism
- Rosiglitazone
- Signal Transduction
- Smad Proteins/genetics
- Smad Proteins/metabolism
- Sterol Esterase/genetics
- Sterol Esterase/metabolism
- Thiazolidinediones/pharmacology
Collapse
Affiliation(s)
- Salvatore Modica
- Swiss Federal Institute of Technology, Department of Health Science, Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology, Schwerzenbach 8603, Switzerland
| | - Leon G Straub
- Swiss Federal Institute of Technology, Department of Health Science, Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology, Schwerzenbach 8603, Switzerland
| | - Miroslav Balaz
- Swiss Federal Institute of Technology, Department of Health Science, Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology, Schwerzenbach 8603, Switzerland
| | - Wenfei Sun
- Swiss Federal Institute of Technology, Department of Health Science, Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology, Schwerzenbach 8603, Switzerland
| | - Lukas Varga
- Obesity section of Diabetes Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine and University Hospital, Comenius University, 811 02 Bratislava, Slovakia
| | - Patrik Stefanicka
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine and University Hospital, Comenius University, 811 02 Bratislava, Slovakia
| | - Milan Profant
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine and University Hospital, Comenius University, 811 02 Bratislava, Slovakia
| | - Eric Simon
- Target Discovery Research, Boehringer Ingelheim Pharma, 88400 Biberach/Riss, Germany
| | - Heike Neubauer
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma, 88400 Biberach/Riss, Germany
| | - Barbara Ukropcova
- Obesity section of Diabetes Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; Institute of Pathophysiology, Faculty of Medicine, Comenius University, 811 02 Bratislava, Slovakia
| | - Jozef Ukropec
- Obesity section of Diabetes Laboratory, Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Christian Wolfrum
- Swiss Federal Institute of Technology, Department of Health Science, Institute of Food Nutrition and Health, Laboratory of Translational Nutrition Biology, Schwerzenbach 8603, Switzerland.
| |
Collapse
|
144
|
Cheng BH, Leng L, Wu MQ, Zhang Q, Zhang XY, Xu SS, Cao ZP, Li YM, Luan P, Li H. Expression analysis of bone morphogenetic protein 4 between fat and lean birds in adipose tissue and serum. Domest Anim Endocrinol 2016; 56:13-9. [PMID: 26945137 DOI: 10.1016/j.domaniend.2016.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/04/2016] [Accepted: 01/23/2016] [Indexed: 01/15/2023]
Abstract
The objectives of the present study were to characterize the tissue expression of chicken (Gallus gallus) bone morphogenetic protein 4 (BMP4) and compare differences in its expression in abdominal fat tissue and serum between fat and lean birds and to determine a potential relationship between the expression of BMP4 and abdominal fat tissue growth and development. The results showed that chicken BMP4 messenger RNA (mRNA) and protein were expressed in various tissues, and the expression levels of BMP4 transcript and protein were relatively higher in adipose tissues. In addition, the mRNA and protein expression levels of BMP4 in abdominal fat tissue of fat males were lower than those of lean males at 1, 2, 5, and 7 wk of age (P < 0.05). Furthermore, the serum BMP4 content of fat males was lower than that of lean males at 7 wk of age (P < 0.05). BMP4 mRNA expression levels were significantly higher in preadipocytes than those in mature adipocytes (P < 0.05), and the expression level decreased during differentiation in vitro (P < 0.05). These results suggested that chicken BMP4 might affect abdominal fat deposition through differences in its expression level. The results of this study will provide basic molecular information for studying the role of BMP4 in the regulation of adipogenesis in avian species.
Collapse
Affiliation(s)
- B H Cheng
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - L Leng
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - M Q Wu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Q Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - X Y Zhang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - S S Xu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Z P Cao
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Y M Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - P Luan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - H Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture, Harbin 150030, Heilongjiang, China; Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, Heilongjiang, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China.
| |
Collapse
|
145
|
Cell Models and Their Application for Studying Adipogenic Differentiation in Relation to Obesity: A Review. Int J Mol Sci 2016; 17:ijms17071040. [PMID: 27376273 PMCID: PMC4964416 DOI: 10.3390/ijms17071040] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 02/08/2023] Open
Abstract
Over the last several years, the increasing prevalence of obesity has favored an intense study of adipose tissue biology and the precise mechanisms involved in adipocyte differentiation and adipogenesis. Adipocyte commitment and differentiation are complex processes, which can be investigated thanks to the development of diverse in vitro cell models and molecular biology techniques that allow for a better understanding of adipogenesis and adipocyte dysfunction associated with obesity. The aim of the present work was to update the different animal and human cell culture models available for studying the in vitro adipogenic differentiation process related to obesity and its co-morbidities. The main characteristics, new protocols, and applications of the cell models used to study the adipogenesis in the last five years have been extensively revised. Moreover, we depict co-cultures and three-dimensional cultures, given their utility to understand the connections between adipocytes and their surrounding cells in adipose tissue.
Collapse
|
146
|
AMP-Activated Kinase (AMPK) Activation by AICAR in Human White Adipocytes Derived from Pericardial White Adipose Tissue Stem Cells Induces a Partial Beige-Like Phenotype. PLoS One 2016; 11:e0157644. [PMID: 27322180 PMCID: PMC4913939 DOI: 10.1371/journal.pone.0157644] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/02/2016] [Indexed: 12/23/2022] Open
Abstract
Beige adipocytes are special cells situated in the white adipose tissue. Beige adipocytes, lacking thermogenic cues, morphologically look quite similar to regular white adipocytes, but with a markedly different response to adrenalin. White adipocytes respond to adrenergic stimuli by enhancing lipolysis, while in beige adipocytes adrenalin induces mitochondrial biogenesis too. A key step in the differentiation and function of beige adipocytes is the deacetylation of peroxisome proliferator-activated receptor (PPARγ) by SIRT1 and the consequent mitochondrial biogenesis. AMP-activated protein kinase (AMPK) is an upstream activator of SIRT1, therefore we set out to investigate the role of AMPK in beige adipocyte differentiation using human adipose-derived mesenchymal stem cells (hADMSCs) from pericardial adipose tissue. hADMSCs were differentiated to white and beige adipocytes and the differentiation medium of the white adipocytes was supplemented with 100 μM [(2R,3S,4R,5R)-5-(4-Carbamoyl-5-aminoimidazol-1-yl)-3,4-dihydroxyoxolan-2-yl]methyl dihydrogen phosphate (AICAR), a known activator of AMPK. The activation of AMPK with AICAR led to the appearance of beige-like morphological properties in differentiated white adipocytes. Namely, smaller lipid droplets appeared in AICAR-treated white adipocytes in a similar fashion as in beige cells. Moreover, in AICAR-treated white adipocytes the mitochondrial network was more fused than in white adipocytes; a fused mitochondrial system was characteristic to beige adipocytes. Despite the morphological similarities between AICAR-treated white adipocytes and beige cells, functionally AICAR-treated white adipocytes were similar to white adipocytes. We were unable to detect increases in basal or cAMP-induced oxygen consumption rate (a marker of mitochondrial biogenesis) when comparing control and AICAR-treated white adipocytes. Similarly, markers of beige adipocytes such as TBX1, UCP1, CIDEA, PRDM16 and TMEM26 remained the same when comparing control and AICAR-treated white adipocytes. Our data point out that in human pericardial hADMSCs the role of AMPK activation in controlling beige differentiation is restricted to morphological features, but not to actual metabolic changes.
Collapse
|
147
|
Pellegrinelli V, Carobbio S, Vidal-Puig A. Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues. Diabetologia 2016; 59:1075-88. [PMID: 27039901 PMCID: PMC4861754 DOI: 10.1007/s00125-016-3933-4] [Citation(s) in RCA: 291] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/23/2016] [Indexed: 02/07/2023]
Abstract
White adipose tissue (WAT) has key metabolic and endocrine functions and plays a role in regulating energy homeostasis and insulin sensitivity. WAT is characterised by its capacity to adapt and expand in response to surplus energy through processes of adipocyte hypertrophy and/or recruitment and proliferation of precursor cells in combination with vascular and extracellular matrix remodelling. However, in the context of sustained obesity, WAT undergoes fibro-inflammation, which compromises its functionality, contributing to increased risk of type 2 diabetes and cardiovascular diseases. Conversely, brown adipose tissue (BAT) and browning of WAT represent potential therapeutic approaches, since dysfunctional white adipocyte-induced lipid overspill can be halted by BAT/browning-mediated oxidative anti-lipotoxic effects. Better understanding of the cellular and molecular pathophysiological mechanisms regulating adipocyte size, number and depot-dependent expansion has become a focus of interest over recent decades. Here, we summarise the mechanisms contributing to adipose tissue (AT) plasticity and function including characteristics and cellular complexity of the various adipose depots and we discuss recent insights into AT origins, identification of adipose precursors, pathophysiological regulation of adipogenesis and its relation to WAT/BAT expandability in obesity and its associated comorbidities.
Collapse
Affiliation(s)
- Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK.
| | - Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Level 4, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK.
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK.
| |
Collapse
|
148
|
Abstract
Bone morphogenetic proteins (BMPs), originally identified as osteoinductive components in extracts derived from bone, are now known to play important roles in a wide array of processes during formation and maintenance of various organs including bone, cartilage, muscle, kidney, and blood vessels. BMPs and the related "growth and differentiation factors" (GDFs) are members of the transforming growth factor β (TGF-β) family, and transduce their signals through type I and type II serine-threonine kinase receptors and their intracellular downstream effectors, including Smad proteins. Furthermore, BMP signals are finely tuned by various agonists and antagonists. Because deregulation of the BMP activity at multiple steps in signal transduction is linked to a wide variety of human diseases, therapeutic use of activators and inhibitors of BMP signaling will provide potential avenues for the treatment of the human disorders that are caused by hypo- and hyperactivation of BMP signals, respectively.
Collapse
Affiliation(s)
- Takenobu Katagiri
- Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical University, Hidaka-shi, Saitama 350-1241, Japan
| | - Tetsuro Watabe
- Section of Biochemistry, Department of Bio-Matrix, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8549, Japan
| |
Collapse
|
149
|
Hu W, Zhang Y, Wang L, Lau CW, Xu J, Luo JY, Gou L, Yao X, Chen ZY, Ma RCW, Tian XY, Huang Y. Bone Morphogenic Protein 4-Smad–Induced Upregulation of Platelet-Derived Growth Factor AA Impairs Endothelial Function. Arterioscler Thromb Vasc Biol 2016; 36:553-60. [DOI: 10.1161/atvbaha.115.306302] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 01/04/2016] [Indexed: 01/29/2023]
Abstract
Objective—
Bone morphogenic protein 4 (BMP4) is an important mediator of endothelial dysfunction in cardio-metabolic diseases, whereas platelet-derived growth factors (PDGFs) are major angiogenic and proinflammatory mediator, although the functional link between these 2 factors is unknown. The present study investigated whether PDGF mediates BMP4-induced endothelial dysfunction in diabetes mellitus.
Approach and Results—
We generated Ad-Bmp4 to overexpress Bmp4 and Ad-Pdgfa-shRNA to knockdown Pdgfa in mice through tail intravenous injection. SMAD4-shRNA lentivirus, SMAD1-shRNA, and SMAD5 shRNA adenovirus were used for knockdown in human and mouse endothelial cells. We found that PDGF-AA impaired endothelium-dependent vasodilation in aortas and mesenteric resistance arteries. BMP4 upregulated PDGF-AA in human and mouse endothelial cells, which was abolished by BMP4 antagonist noggin or knockdown of SMAD1/5 or SMAD4. BMP4-impared relaxation in mouse aorta was also ameliorated by PDGF-AA neutralizing antibody. Tail injection of Ad-Pdgfa-shRNA ameliorates endothelial dysfunction induced by Bmp4 overexpression (Ad-Bmp4) in vivo. Serum PDGF-AA was elevated in both diabetic patients and diabetic
db/db
mice compared with nondiabetic controls. Pdgfa-shRNA or Bmp4-shRNA adenovirus reduced serum PDGF-AA concentration in
db/db
mice. PDGF-AA neutralizing antibody or tail injection with Pdgfa-shRNA adenovirus improved endothelial function in aortas and mesenteric resistance arteries from
db/db
mice. The effect of PDGF-AA on endothelial function in mouse aorta was also inhibited by Ad-Pdgfra-shRNA to inhibit PDGFRα.
Conclusions—
The present study provides novel evidences to show that PDGF-AA impairs endothelium-dependent vasodilation and PDGF-AA mediates BMP4-induced adverse effect on endothelial cell function through SMAD1/5- and SMAD4-dependent mechanisms. Inhibition of PGDF-AA ameliorates vascular dysfunction in diabetic mice.
Collapse
Affiliation(s)
- Weining Hu
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Zhang
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Li Wang
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Wai Lau
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jian Xu
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiang-Yun Luo
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lingshan Gou
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaoqiang Yao
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhen-Yu Chen
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald Ching Wan Ma
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao Yu Tian
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- From the Shenzhen Research Institute, School of Biomedical Sciences, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences (W.H., Y.Z., L.W., C.W.L., J.X., J.-Y.L., L.G., X.Y., X.Y.T., Y.H), the School of Life Science (Z.-Y.C.), Department of Medicine and Therapeutics, the Prince of Wales Hospital, Hong Kong; and Institute of Diabetes and Obesity (R.C.W.M), Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
150
|
Tencerova M, Kassem M. The Bone Marrow-Derived Stromal Cells: Commitment and Regulation of Adipogenesis. Front Endocrinol (Lausanne) 2016; 7:127. [PMID: 27708616 PMCID: PMC5030474 DOI: 10.3389/fendo.2016.00127] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/05/2016] [Indexed: 12/13/2022] Open
Abstract
Bone marrow (BM) microenvironment represents an important compartment of bone that regulates bone homeostasis and the balance between bone formation and bone resorption depending on the physiological needs of the organism. Abnormalities of BM microenvironmental dynamics can lead to metabolic bone diseases. BM stromal cells (also known as skeletal or mesenchymal stem cells) [bone marrow stromal stem cell (BMSC)] are multipotent stem cells located within BM stroma and give rise to osteoblasts and adipocytes. However, cellular and molecular mechanisms of BMSC lineage commitment to adipocytic lineage and regulation of BM adipocyte formation are not fully understood. In this review, we will discuss recent findings pertaining to identification and characterization of adipocyte progenitor cells in BM and the regulation of differentiation into mature adipocytes. We have also emphasized the clinical relevance of these findings.
Collapse
Affiliation(s)
- Michaela Tencerova
- Department of Molecular Endocrinology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Danish Diabetes Academy, Novo Nordisk Foundation, Odense, Denmark
- *Correspondence: Michaela Tencerova,
| | - Moustapha Kassem
- Department of Molecular Endocrinology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Danish Diabetes Academy, Novo Nordisk Foundation, Odense, Denmark
- Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|