751
|
Rosenbaum M, Knight R, Leibel RL. The gut microbiota in human energy homeostasis and obesity. Trends Endocrinol Metab 2015; 26:493-501. [PMID: 26257300 PMCID: PMC4862197 DOI: 10.1016/j.tem.2015.07.002] [Citation(s) in RCA: 293] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/03/2015] [Accepted: 07/10/2015] [Indexed: 12/15/2022]
Abstract
Numerous studies of rodents suggest that the gut microbiota populations are sensitive to genetic and environmental influences, and can produce or influence afferent signals that directly or indirectly impinge on energy homeostatic systems affecting both energy balance (weight gain or loss) and energy stores. Fecal transplants from obese and lean human, and from mouse donors to gnotobiotic mice, result in adoption of the donor somatotype by the formerly germ-free rodents. Thus, the microbiota is certainly implicated in the development of obesity, adiposity-related comorbidities, and the response to interventions designed to achieve sustained weight reduction in mice. More studies are needed to determine whether the microbiota plays a similarly potent role in human body-weight regulation and obesity.
Collapse
Affiliation(s)
- Michael Rosenbaum
- Columbia University, Department of Pediatrics, Division of Molecular Genetics, New York, NY 10032, USA.
| | - Rob Knight
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Rudolph L Leibel
- Columbia University, Department of Pediatrics, Division of Molecular Genetics, New York, NY 10032, USA
| |
Collapse
|
752
|
Roberts-Toler C, O’Neill BT, Cypess AM. Diet-induced obesity causes insulin resistance in mouse brown adipose tissue. Obesity (Silver Spring) 2015; 23:1765-70. [PMID: 26242777 PMCID: PMC4551605 DOI: 10.1002/oby.21134] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/22/2015] [Accepted: 04/06/2015] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Diet-induced obesity (DIO) causes several pathophysiological changes in adipose tissue. Increased inflammation reduces white adipose tissue (WAT) insulin sensitivity and contributes to the development of diabetes. However, little is known about how DIO alters the function of brown adipose tissue (BAT), an organ that consumes calories by β3-adrenergic receptor (AR)-mediated thermogenesis and helps regulate energy balance. METHODS To test the effects of DIO on BAT, we fed 6-week-old C57BL/6 mice either a normal chow diet (NCD) or a high-fat diet (HFD). After 16 additional weeks, we measured body fat, WAT, and BAT mRNA expression, glucose tolerance, and rates of glucose uptake in response to insulin and the β3-AR agonist mirabegron. RESULTS Compared with NCD, HFD increased body fat and impaired glucose tolerance. Both WAT and BAT had higher mRNA levels of markers of inflammation, including TNFα and F4/80. Insulin signaling in BAT and WAT was reduced, with decreased Akt phosphorylation. Diet-normalized BAT glucose uptake rates were lower in response to mirabegron. CONCLUSIONS These results support a model in which DIO leads to BAT inflammation and insulin resistance, leading to a broader impairment of BAT function.
Collapse
Affiliation(s)
- Carla Roberts-Toler
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Brian T. O’Neill
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Aaron M. Cypess
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
- Translational Physiology Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
753
|
Abstract
A number of chronic metabolic pathologies, including obesity, diabetes, cardiovascular disease, asthma, and cancer, cluster together to present the greatest threat to human health. As research in this field has advanced, it has become clear that unresolved metabolic inflammation, organelle dysfunction, and other cellular and metabolic stresses underlie the development of these chronic metabolic diseases. However, the relationship between these systems and pathological mechanisms is poorly understood. Here we discuss the role of cellular Ca(2+) homeostasis as a critical mechanism integrating the myriad of cellular and subcellular dysfunctional networks found in metabolic tissues such as liver and adipose tissue in the context of metabolic disease, particularly in obesity and diabetes.
Collapse
|
754
|
Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation. Immunity 2015; 42:1005-19. [PMID: 26084021 DOI: 10.1016/j.immuni.2015.06.006] [Citation(s) in RCA: 451] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-33 (IL-33) is a nuclear-associated cytokine of the IL-1 family originally described as a potent inducer of allergic type 2 immunity. IL-33 signals via the receptor ST2, which is highly expressed on group 2 innate lymphoid cells (ILC2s) and T helper 2 (Th2) cells, thus underpinning its association with helminth infection and allergic pathology. Recent studies have revealed ST2 expression on subsets of regulatory T cells, and for a role for IL-33 in tissue homeostasis and repair that suggests previously unrecognized interactions within these cellular networks. IL-33 can participate in pathologic fibrotic reactions, or, in the setting of microbial invasion, can cooperate with inflammatory cytokines to promote responses by cytotoxic NK cells, Th1 cells, and CD8(+) T cells. Here, we highlight the regulation and function of IL-33 and ST2 and review their roles in homeostasis, damage, and inflammation, suggesting a conceptual framework for future studies.
Collapse
Affiliation(s)
- Ari B Molofsky
- Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA; Department of Laboratory Medicine, University of California, San Francisco, 94143-0795, USA
| | - Adam K Savage
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA
| | - Richard M Locksley
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Medicine, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA.
| |
Collapse
|
755
|
Lettieri Barbato D, Tatulli G, Maria Cannata S, Bernardini S, Aquilano K, Ciriolo MR. Glutathione Decrement Drives Thermogenic Program In Adipose Cells. Sci Rep 2015; 5:13091. [PMID: 26260892 PMCID: PMC4531326 DOI: 10.1038/srep13091] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/17/2015] [Indexed: 01/21/2023] Open
Abstract
Adipose tissue metabolically adapts to external stimuli. We demonstrate that the induction of the thermogenic program in white adipocytes, through cold exposure in mice or in vitro adrenergic stimulation, is accompanied by a decrease in the intracellular content of glutathione (GSH). Moreover, the treatment with a GSH depleting agent, buthionine sulfoximine (BSO), recapitulates the effect of cold exposure resulting in the induction of thermogenic program. In particular, BSO treatment leads to enhanced uncoupling respiration as demonstrated by increased expression of thermogenic genes (e.g. Ucp1, Ppargc1a), augmented oxygen consumption and decreased mitochondrial transmembrane potential. Buffering GSH decrement by pre-treatment with GSH ester prevents the up-regulation of typical markers of uncoupling respiration. We demonstrate that FoxO1 activation is responsible for the conversion of white adipocytes into a brown phenotype as the “browning” effects of BSO are completely abrogated in cells down-regulating FoxO1. In mice, the BSO-mediated up-regulation of uncoupling genes results in weight loss that is at least in part ascribed to adipose tissue mass reduction. The induction of thermogenic program has been largely proposed to counteract obesity-related diseases. Based on these findings, we propose GSH as a novel therapeutic target to increase energy expenditure in adipocytes.
Collapse
Affiliation(s)
- Daniele Lettieri Barbato
- Dept. Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Giuseppe Tatulli
- Scientific Institute for Research Hospitalization and Health Care and Università Telematica San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Stefano Maria Cannata
- Dept. Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Sergio Bernardini
- Dept. Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Katia Aquilano
- 1] Dept. Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy [2] Scientific Institute for Research Hospitalization and Health Care and Università Telematica San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Maria R Ciriolo
- 1] Dept. Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy [2] Scientific Institute for Research Hospitalization and Health Care and Università Telematica San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| |
Collapse
|
756
|
Of mice and men: novel insights regarding constitutive and recruitable brown adipocytes. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2015; 5:S15-20. [PMID: 27152169 DOI: 10.1038/ijosup.2015.5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 02/12/2015] [Indexed: 01/02/2023]
Abstract
Recently, there has been great attention given to the possibility of combating obesity by targeting brown fat activity or increasing differentiation of brown adipocytes in white fat depots through a process termed 'browning'. Sympathetic innervation of brown and white adipose tissues provides adrenergic input that drives thermogenesis and regulates fatty acid metabolism, as well as stimulating adipogenesis of recruitable brown adipocyte tissue (rBAT, also known as beige or brite) in white fat. Other factors acting in an endocrine or autocrine/paracrine manner in adipose tissue may also stimulate browning. There have been significant recent advances in understanding the mechanisms of increasing adipose tissue energy expenditure, as well as how brown adipocytes appear in white fat depots, including via de novo adipogenesis from tissue precursor cells. In this article, we integrate this new knowledge with a historical perspective on the discovery of 'browning'. We also provide an overview of constitutive BAT vs rBAT in mouse and human.
Collapse
|
757
|
Morano S, Romagnoli E, Filardi T, Nieddu L, Mandosi E, Fallarino M, Turinese I, Dagostino MP, Lenzi A, Carnevale V. Short-term effects of glucagon-like peptide 1 (GLP-1) receptor agonists on fat distribution in patients with type 2 diabetes mellitus: an ultrasonography study. Acta Diabetol 2015; 52:727-32. [PMID: 25577244 DOI: 10.1007/s00592-014-0710-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/26/2014] [Indexed: 01/22/2023]
Abstract
AIMS Glucagon-like peptide 1 receptor agonists (GLP-1 RA) induce weight loss and reduction in adipose tissue, but the effects of GLP-1 RA on the distribution of fat deposits have been poorly investigated. METHODS In 25 patients with type 2 diabetes (16 females and 9 males, mean age 63.5 ± 8.8 years), treated with GLP-1 RA (exenatide, n. 12; liraglutide, n.13), both before and 3 months after starting treatment, an abdominal ultrasonographic scan, with Doppler of renal arteries, and echocardiography were performed. Subcutaneous fat width (peri-umbilical and sub-xiphoid), deep fat deposits (pre-aortic, peri-renal, and epicardial), and renal resistive index (RI) were evaluated. RESULTS GLP-1 RA induced highly significant (p < 0.001) decrease in BMI and in fat thickness at all the assessed sites, without differences between exenatide and liraglutide treatment. A slight decrease in RI (p = 0.055) was also found. The percent changes of fat thickness was different between sites (p < 0.025), and the changes in subcutaneous deposits showed no significant correlation (p = 0.064) with those of deep fat deposits. CONCLUSIONS A short course of treatment with GLP-1 RA, besides weight loss, induces a redistribution of adipose tissue deposits, possibly contributing to a better cardiovascular risk profile in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Susanna Morano
- Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
758
|
Ko HJ, Lo CY, Wang BJ, Chiou RYY, Lin SM. Theaflavin-3,3′-digallate, a black tea polyphenol, stimulates lipolysis associated with the induction of mitochondrial uncoupling proteins and AMPK–FoxO3A–MnSOD pathway in 3T3-L1 adipocytes. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.05.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
759
|
Duran ID, Gülçelik NE, Ünal M, Topçuoğlu C, Sezer S, Tuna MM, Berker D, Güler S. Irisin levels in the progression of diabetes in sedentary women. Clin Biochem 2015; 48:1268-72. [PMID: 26234637 DOI: 10.1016/j.clinbiochem.2015.07.098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022]
Abstract
CONTEXT The recently discovered peptide irisin has been hypothesized to be a regulator of body metabolism. However, studies ended up with controversial results. In the present study, we aimed to investigate irisin levels in sedentary women at different stages of prediabetes. DESIGN, SETTING, AND SUBJECTS We performed a cross-sectional analysis of circulating levels of irisin in 263 females similar for age and body mass index (BMI) and the groups included 52 normal glucose tolerance (NGT), 60 isolated impaired fasting glucose (IFG), 36 isolated impaired glucose tolerance (IGT), 65 both IFG and IGT and 50 type 2 diabetic patients. All patients were exercising less than 150 min/week. RESULTS Plasma irisin levels were significantly lower in IFG+IGT (2.86 ± 0.6 μg/mL, p: 0.019) and T2DM (2.83 ± 0,5 μg/mL, p: 0.005) patients compared to NGT (3.16 ± 0.3 μg/mL) patients. After age adjustment there was a negative correlation between irisin and BMI (r: -0.141; p: 0.031), postprandial glucose (PPG) (r: -0.142; p: 0.030), low density lipoprotein-cholesterol (LDL-C) (r: -0.138; p: 0.035) and triglyceride (TG) (r: -0.214; p: 0.001) and a positive correlation between irisin and high density lipoprotein-cholesterol (HDL-C) (r:.142; p: 0.030). After adjustment for age and BMI; PPG (r: -0. 137; p: 0.037), LDL-C (r: -0. 143; p: 0.029) and TG (r: -0.203; p: 0.002) were considered to correlate with irisin levels. Subgroup analysis revealed that TG levels were correlated with irisin levels in IFG (r: -0.347; p: 0.014) and IGT (r: -0.397; p: 0.030) patients. CONCLUSION In our cohort of sedentary women, irisin levels were lower in patients with IFG+IGT and with diabetes than in patients with NGT. There is no correlation between irisin levels and BMI. Irisin is a myokine decreasing gradually with the progression of glucose intolerance and T2DM and is not correlated with BMI in sedentary women.
Collapse
Affiliation(s)
- Iffet Dağdelen Duran
- Ankara Numune Education and Research Hospital, Department of Endocrinology and Metabolic Diseases, Turkey.
| | - Neşe Ersöz Gülçelik
- Ankara Numune Education and Research Hospital, Department of Endocrinology and Metabolic Diseases, Turkey
| | - Mustafa Ünal
- Ankara Numune Education and Research Hospital, Department of Endocrinology and Metabolic Diseases, Turkey
| | - Canan Topçuoğlu
- Ankara Numune Education and Research Hospital, Department of Clinical Biochemistry, Turkey
| | - Sevilay Sezer
- Ankara Numune Education and Research Hospital, Department of Clinical Biochemistry, Turkey
| | - Mazhar Müslüm Tuna
- Ankara Numune Education and Research Hospital, Department of Endocrinology and Metabolic Diseases, Turkey
| | - Dilek Berker
- Ankara Numune Education and Research Hospital, Department of Endocrinology and Metabolic Diseases, Turkey
| | - Serdar Güler
- Ankara Numune Education and Research Hospital, Department of Endocrinology and Metabolic Diseases, Turkey
| |
Collapse
|
760
|
Abstract
Thermogenesis, the production of heat energy, in brown adipose tissue is a significant component of the homeostatic repertoire to maintain body temperature during the challenge of low environmental temperature in many species from mouse to man and plays a key role in elevating body temperature during the febrile response to infection. The sympathetic neural outflow determining brown adipose tissue (BAT) thermogenesis is regulated by neural networks in the CNS which increase BAT sympathetic nerve activity in response to cutaneous and deep body thermoreceptor signals. Many behavioral states, including wakefulness, immunologic responses, and stress, are characterized by elevations in core body temperature to which central command-driven BAT activation makes a significant contribution. Since energy consumption during BAT thermogenesis involves oxidation of lipid and glucose fuel molecules, the CNS network driving cold-defensive and behavioral state-related BAT activation is strongly influenced by signals reflecting the short- and long-term availability of the fuel molecules essential for BAT metabolism and, in turn, the regulation of BAT thermogenesis in response to metabolic signals can contribute to energy balance, regulation of body adipose stores and glucose utilization. This review summarizes our understanding of the functional organization and neurochemical influences within the CNS networks that modulate the level of BAT sympathetic nerve activity to produce the thermoregulatory and metabolic alterations in BAT thermogenesis and BAT energy expenditure that contribute to overall energy homeostasis and the autonomic support of behavior.
Collapse
Affiliation(s)
- Shaun F Morrison
- Department of Neurological Surgery, Oregon Health & Science University, Portland, Oregon
| | | |
Collapse
|
761
|
Kim B, Do MS, Hyun CK. B-cell-activating factor deficiency attenuates high-fat diet-induced glucose intolerance by potentiating adipose tissue function. Biochem Biophys Res Commun 2015. [PMID: 26208451 DOI: 10.1016/j.bbrc.2015.07.099] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
B-cell-activating factor (BAFF) has recently been demonstrated to be expressed in adipocytes and up-regulated by high-fat diet feeding, indicating a possible role in metabolic regulation. Here we show that glucose tolerance was significantly improved in high-fat diet-fed BAFF knockout (BAFF(-/-)) mice. BAFF(-/-) mice revealed higher levels of glucose transporter expression and insulin-stimulated Akt phosphorylation in brown adipose tissue compared to wild type controls. Expression levels of mitochondrial ND5 and genes involved in lipid metabolism were significantly elevated in brown adipose tissue of BAFF(-/-) mice, and this enhancement was found to be mediated by FGF21 and leptin. It was also observed that expression of IL-10 and foxp3 was increased in adipose tissues, as well as PPARγ activity in white adipose tissue. Our findings suggest that suppression of BAFF could have a therapeutic potential for prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Bobae Kim
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea
| | - Myoung-Sool Do
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea
| | - Chang-Kee Hyun
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea.
| |
Collapse
|
762
|
López-Yoldi M, Moreno-Aliaga MJ, Bustos M. Cardiotrophin-1: A multifaceted cytokine. Cytokine Growth Factor Rev 2015; 26:523-32. [PMID: 26188636 DOI: 10.1016/j.cytogfr.2015.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
Cardiotrophin-1 (CT-1) is a member of the gp130 family of cytokines that have pleiotropic functions on different tissues and cell types. Although many effects of CT-1 have been described on the heart, there is an extensive research showing important protective effects in other organs such as liver, kidney or nervous system. Recently, several studies have pointed out that CT-1 might also play a key role in the regulation of body weight and intermediate metabolism. This paper will review many aspects of CT-1 physiological role in several organs and discuss data for consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Matilde Bustos
- Area of Hepatology and Gene Therapy, CIMA (Center for Applied Medical Research) University of Navarra, Pamplona, Spain.
| |
Collapse
|
763
|
Li J, Papadopoulos V. Translocator protein (18 kDa) as a pharmacological target in adipocytes to regulate glucose homeostasis. Biochem Pharmacol 2015; 97:99-110. [PMID: 26123521 DOI: 10.1016/j.bcp.2015.06.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 06/22/2015] [Indexed: 11/28/2022]
Abstract
As a major regulator in obesity and its associated metabolic complications, the proper functioning of adipocytes is crucial for health maintenance, thus serving as an important target for the development of anti-obese and anti-diabetic therapies. There is increasing evidence that mitochondrial malfunction is a pivotal event in disturbing adipocyte cell homeostasis. Among major mitochondrial structure components, the high-affinity drug- and cholesterol-binding outer mitochondrial membrane translocator protein (18 kDa; TSPO) has shown importance across a broad spectrum of mitochondrial functions. Recent studies demonstrated the presence of TSPO in white adipocyte mitochondria of mice, and administration of TSPO drug ligands to obese mice reduced weight gain and lowered glucose level. Therefore, it is of great interest to assess whether TSPO in adipocytes could serve as a drug target to regulate adipocyte activities with potential influence on weight control and glucose metabolism. Two structurally distinct TSPO drug ligands, PK 11195 and FGIN-1-27, improved the intracellular dynamics of 3T3-L1 adipocytes, such as the production and release of adipokines, glucose uptake, and adipogenesis. TSPO knockdown in either differentiated adipocytes or preadipocytes impaired these functions. Findings from 3T3-L1 cells were related to human primary cells, where TSPO expression was tightly associated with the metabolic state of primary adipocytes and the differentiation of primary preadipocytes. These results suggest that TSPO expression is essential to safeguard healthy adipocyte functions, and that TSPO activation in adipocytes improves their metabolic status in regulating glucose homeostasis. Adipocyte TSPO may serve as a pharmacologic target for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Jiehan Li
- Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Vassilios Papadopoulos
- Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada; Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada; Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada.
| |
Collapse
|
764
|
Straub L, Wolfrum C. FGF21, energy expenditure and weight loss - How much brown fat do you need? Mol Metab 2015; 4:605-9. [PMID: 26413466 PMCID: PMC4563019 DOI: 10.1016/j.molmet.2015.06.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/16/2022] Open
Abstract
Background Fibroblast growth factor 21 (FGF21) belongs to the large family of fibroblast growth factors (FGFs). Even though FGF signaling has been mainly implicated in developmental processes, recent studies have demonstrated that FGF21 is an important regulator of whole body energy expenditure and metabolism, in obesity. Scope of review Given the fact that obesity has developed epidemic proportions, not just in industrialized countries, FGF21 has emerged as a novel therapeutic avenue to treat obesity as well as associated metabolic disorders. While the metabolic effects of FGF21 are undisputed, the mechanisms by which FGF21 regulate weight loss have not yet been fully resolved. Until recently it was believed that FGF21 induces brown fat activity, thereby enhancing energy expenditure, which concomitantly leads to weight loss. Novel studies have challenged this concept as they could demonstrate that a part of the FGF21 mediated effects are retained in a mouse model of impaired brown adipose tissue function. Major conclusions The review illustrates the recent advances in FGF21 research and discusses the role of FGF21 in the regulation of energy expenditure linked to brown fat activity.
Collapse
Affiliation(s)
- Leon Straub
- Swiss Federal Institute of Technology, ETH Zürich, Institute of Food Nutrition and Health, Schorenstr. 16, 8603 Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Swiss Federal Institute of Technology, ETH Zürich, Institute of Food Nutrition and Health, Schorenstr. 16, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
765
|
Adipokines influence the inflammatory balance in autoimmunity. Cytokine 2015; 75:272-9. [PMID: 26044595 DOI: 10.1016/j.cyto.2015.04.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 03/16/2015] [Accepted: 04/12/2015] [Indexed: 01/04/2023]
Abstract
Over the past few decades, our understanding of the role of adipose tissue has changed dramatically. Far from simply being a site of energy storage or a modulator of the endocrine system, adipose tissue has emerged as an important regulator of multiple important processes including inflammation. Adipokines are a diverse family of soluble mediators with a range of specific actions on the immune response. Autoimmune diseases are perpetuated by chronic inflammatory responses but the exact etiology of these diseases remains elusive. While researchers continue to investigate these causes, millions of people continue to suffer from chronic diseases. To this end, an increased interest has developed in the connection between adipose tissue-secreted proteins that influence inflammation and the onset and perpetuation of autoimmunity. This review will focus on recent advances in adipokine research with specific attention on a subset of adipokines that have been associated with autoimmune diseases.
Collapse
|
766
|
Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell 2015; 161:146-160. [PMID: 25815992 DOI: 10.1016/j.cell.2015.02.022] [Citation(s) in RCA: 339] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 02/07/2023]
Abstract
Obesity is an increasingly prevalent disease worldwide. While genetic and environmental factors are known to regulate the development of obesity and associated metabolic diseases, emerging studies indicate that innate and adaptive immune cell responses in adipose tissue have critical roles in the regulation of metabolic homeostasis. In the lean state, type 2 cytokine-associated immune cell responses predominate in white adipose tissue and protect against weight gain and insulin resistance through direct effects on adipocytes and elicitation of beige adipose. In obesity, these metabolically beneficial immune pathways become dysregulated, and adipocytes and other factors initiate metabolically deleterious type 1 inflammation that impairs glucose metabolism. This review discusses our current understanding of the functions of different types of adipose tissue and how immune cells regulate adipocyte function and metabolic homeostasis in the context of health and disease and highlights. We also highlight the potential of targeting immuno-metabolic pathways as a therapeutic strategy to treat obesity and associated diseases.
Collapse
Affiliation(s)
- Jonathan R Brestoff
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, 10021, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, 10021, USA.
| |
Collapse
|
767
|
Hoffmann LS, Etzrodt J, Willkomm L, Sanyal A, Scheja L, Fischer AWC, Stasch JP, Bloch W, Friebe A, Heeren J, Pfeifer A. Stimulation of soluble guanylyl cyclase protects against obesity by recruiting brown adipose tissue. Nat Commun 2015; 6:7235. [PMID: 26011238 PMCID: PMC4455111 DOI: 10.1038/ncomms8235] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/21/2015] [Indexed: 12/31/2022] Open
Abstract
Obesity is characterized by a positive energy balance and expansion of white adipose tissue (WAT). In contrast, brown adipose tissue (BAT) combusts energy to produce heat. Here we show that a small molecule stimulator (BAY 41-8543) of soluble guanylyl cyclase (sGC), which produces the second messenger cyclic GMP (cGMP), protects against diet-induced weight gain, induces weight loss in established obesity, and also improves the diabetic phenotype. Mechanistically, the haeme-dependent sGC stimulator BAY 41-8543 enhances lipid uptake into BAT and increases whole-body energy expenditure, whereas ablation of the haeme-containing β1-subunit of sGC severely impairs BAT function. Notably, the sGC stimulator enhances differentiation of human brown adipocytes as well as induces 'browning' of primary white adipocytes. Taken together, our data suggest that sGC is a potential pharmacological target for the treatment of obesity and its comorbidities.
Collapse
Affiliation(s)
- Linda S Hoffmann
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany
| | - Jennifer Etzrodt
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany
| | - Lena Willkomm
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Institute of Cardiovascular Research and Sports Medicine, Cologne D-50735, Germany
| | - Abhishek Sanyal
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany.,Research Training Group 1873, University of Bonn, Bonn D-53127, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Alexander W C Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Johannes-Peter Stasch
- Bayer Pharma AG, Wuppertal D-42113, Germany.,Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle an der Saale D-06120, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Institute of Cardiovascular Research and Sports Medicine, Cologne D-50735, Germany
| | - Andreas Friebe
- Institute of Physiology, Martin Luther University Würzburg, Würzburg D-97070, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, Bonn D-53105, Germany.,Research Training Group 1873, University of Bonn, Bonn D-53127, Germany.,PharmaCenter, University of Bonn, Bonn D-53119, Germany
| |
Collapse
|
768
|
Dallinga-Thie GM, Nieuwdorp M. GLP1, an Important Regulator of Intestinal Lipid Metabolism. Arterioscler Thromb Vasc Biol 2015; 35:1048-9. [DOI: 10.1161/atvbaha.115.305479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Geesje M. Dallinga-Thie
- From the Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands (G.M.D.-T., M.N.); Department of Internal Medicine, VUMC Diabetes Center, VUMC, Amsterdam, The Netherlands (M.N.); and Wallenberg Laboratory, University of Gothenberg, Gothenberg, Sweden (M.N.)
| | - Max Nieuwdorp
- From the Departments of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands (G.M.D.-T., M.N.); Department of Internal Medicine, VUMC Diabetes Center, VUMC, Amsterdam, The Netherlands (M.N.); and Wallenberg Laboratory, University of Gothenberg, Gothenberg, Sweden (M.N.)
| |
Collapse
|
769
|
Wang GX, Zhao XY, Lin JD. The brown fat secretome: metabolic functions beyond thermogenesis. Trends Endocrinol Metab 2015; 26:231-7. [PMID: 25843910 PMCID: PMC4417028 DOI: 10.1016/j.tem.2015.03.002] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/08/2015] [Accepted: 03/11/2015] [Indexed: 12/29/2022]
Abstract
Brown fat is highly active in fuel oxidation and dissipates chemical energy through uncoupling protein (UCP)1-mediated heat production. Activation of brown fat leads to increased energy expenditure, reduced adiposity, and lower plasma glucose and lipid levels, thus contributing to better homeostasis. Uncoupled respiration and thermogenesis have been considered to be responsible for the metabolic benefits of brown adipose tissue. Recent studies have demonstrated that brown adipocytes also secrete factors that act locally and systemically to influence fuel and energy metabolism. This review discusses the evidence supporting a thermogenesis-independent role of brown fat, particularly through its release of secreted factors, and their implications in physiology and therapeutic development.
Collapse
Affiliation(s)
- Guo-Xiao Wang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Xu-Yun Zhao
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA.
| |
Collapse
|
770
|
Than A, He HL, Chua SH, Xu D, Sun L, Leow MKS, Chen P. Apelin Enhances Brown Adipogenesis and Browning of White Adipocytes. J Biol Chem 2015; 290:14679-91. [PMID: 25931124 DOI: 10.1074/jbc.m115.643817] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Indexed: 12/17/2022] Open
Abstract
Brown adipose tissue expends energy in the form of heat via the mitochondrial uncoupling protein UCP1. Recent studies showed that brown adipose tissue is present in adult humans and may be exploited for its anti-obesity and anti-diabetes actions. Apelin is an adipocyte-derived hormone that plays important roles in energy metabolism. Here, we report that apelin-APJ signaling promotes brown adipocyte differentiation by increasing the expressions of brown adipogenic and thermogenic transcriptional factors via the PI3K/Akt and AMPK signaling pathways. It is also found that apelin relieves the TNFα inhibition on brown adipogenesis. In addition, apelin increases the basal activity of brown adipocytes, as evidenced by the increased PGC1α and UCP1 expressions, mitochondrial biogenesis, and oxygen consumption. Finally, we provide both in vitro and in vivo evidence that apelin is able to increase the brown-like characteristics in white adipocytes. This study, for the first time, reveals the brown adipogenic and browning effects of apelin and suggests a potential therapeutic route to combat obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Aung Than
- From the Bioengineering Program, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Hui Ling He
- From the Bioengineering Program, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Si Hui Chua
- From the Bioengineering Program, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Dan Xu
- the Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, and
| | - Lei Sun
- the Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, and
| | - Melvin Khee-Shing Leow
- the Endocrine and Diabetes Clinic, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Peng Chen
- From the Bioengineering Program, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457,
| |
Collapse
|
771
|
Schade KN, Baranwal A, Liang C, Mirbolooki MR, Mukherjee J. Preliminary evaluation of β3-adrenoceptor agonist-induced 18F-FDG metabolic activity of brown adipose tissue in obese Zucker rat. Nucl Med Biol 2015; 42:691-4. [PMID: 25934347 DOI: 10.1016/j.nucmedbio.2015.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/07/2015] [Accepted: 04/10/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND We have investigated β3-adrenoceptor agonist mediated brown adipose tissue (BAT) activation using (18)F-FDG PET/CT in Zucker lean (ZL) and obese (ZF) rats. METHODS (18)F-FDG was injected into ZL and ZF rats pretreated with saline or agonist CL316,243 for scans. (18)F-FDG metabolic activity was computed as standard uptake values. RESULTS CL316,243 in ZL activated BAT up to 4-fold compared to saline, while ZF BAT was only up by 2 fold. The decreased activation was consistent with lower β3-adrenoceptor levels in ZF rats. CONCLUSIONS The genetically modified ZL and ZF rats may provide a useful rat model to evaluate the significance of β3-adrenoceptor agonist-induced BAT activation in obesity.
Collapse
Affiliation(s)
- Kimberly N Schade
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697
| | - Aparna Baranwal
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697
| | - Christopher Liang
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697
| | - M Reza Mirbolooki
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697
| | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697.
| |
Collapse
|
772
|
Gonzalez B, Forcales SV, Perucho M. Second German-Catalan workshop on epigenetics & cancer. Epigenetics 2015; 10:352-9. [PMID: 25849957 DOI: 10.1080/15592294.2015.1023499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The Second German-Catalan Workshop on Epigenetics and Cancer was held in Barcelona on November 19-21, 2014. The workshop brought together, for the second time, scientists from 2 German and 2 Catalan research institutions: the DKFZ, from Heidelberg, the CRCME, from Freiburg, and the IMPPC and PEBC/IDIBELL, both from Barcelona. The German-Catalan Workshops are intended to establish the framework for building a Research School to foster collaborations between researchers from the different institutions. Exchange programs for graduate students are among the activities of the future School. The topics presented and discussed in 33 talks were diverse and included work on DNA methylation, histone modifications, chromatin biology, characterization of imprinted regions in human tissues, non-coding RNAs, and epigenetic drug discovery. Among novel developments from the previous Workshop are the report of the epigenetics angle of the Warburg effect and the long-range trans-acting interaction of DNA methylation and of nucleosome remodeling. A shift in the view on DNA methylation became apparent by the realization of the intertwined interplay between hyper- and hypo-methylation in differentiation and cancer.
Collapse
Affiliation(s)
- Beatriz Gonzalez
- a Institute of Predictive and Personalized Medicine of Cancer (IMPPC); Campus Can Ruti ; Badalona , Barcelona , Spain
| | | | | |
Collapse
|
773
|
Dodd GT, Decherf S, Loh K, Simonds SE, Wiede F, Balland E, Merry TL, Münzberg H, Zhang ZY, Kahn BB, Neel BG, Bence KK, Andrews ZB, Cowley MA, Tiganis T. Leptin and insulin act on POMC neurons to promote the browning of white fat. Cell 2015; 160:88-104. [PMID: 25594176 DOI: 10.1016/j.cell.2014.12.022] [Citation(s) in RCA: 280] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 10/14/2014] [Accepted: 12/10/2014] [Indexed: 01/06/2023]
Abstract
The primary task of white adipose tissue (WAT) is the storage of lipids. However, "beige" adipocytes also exist in WAT. Beige adipocytes burn fat and dissipate the energy as heat, but their abundance is diminished in obesity. Stimulating beige adipocyte development, or WAT browning, increases energy expenditure and holds potential for combating metabolic disease and obesity. Here, we report that insulin and leptin act together on hypothalamic neurons to promote WAT browning and weight loss. Deletion of the phosphatases PTP1B and TCPTP enhanced insulin and leptin signaling in proopiomelanocortin neurons and prevented diet-induced obesity by increasing WAT browning and energy expenditure. The coinfusion of insulin plus leptin into the CNS or the activation of proopiomelanocortin neurons also increased WAT browning and decreased adiposity. Our findings identify a homeostatic mechanism for coordinating the status of energy stores, as relayed by insulin and leptin, with the central control of WAT browning.
Collapse
Affiliation(s)
- Garron T Dodd
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Stephanie Decherf
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Kim Loh
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | | | - Florian Wiede
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Eglantine Balland
- Department of Physiology, Monash University, Victoria 3800, Australia
| | - Troy L Merry
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Heike Münzberg
- Pennington Biomedical Research Center, LSU Systems, Baton Rouge, LA 70808, USA
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Benjamin G Neel
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital and Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Kendra K Bence
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zane B Andrews
- Department of Physiology, Monash University, Victoria 3800, Australia
| | - Michael A Cowley
- Department of Physiology, Monash University, Victoria 3800, Australia
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia.
| |
Collapse
|
774
|
Hu X, Luo P, Peng X, Song T, Zhou Y, Wei H, Peng J, Jiang S. Molecular cloning, expression pattern analysis of porcine Rb1 gene and its regulatory roles during primary dedifferentiated fat cells adipogenic differentiation. Gen Comp Endocrinol 2015; 214:77-86. [PMID: 25626122 DOI: 10.1016/j.ygcen.2015.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 01/13/2015] [Accepted: 01/19/2015] [Indexed: 11/19/2022]
Abstract
Adipocytes are the main constituent of adipose tissue and are considered to be a corner stone in the homeostatic control of whole body metabolism. Recent reports evidenced that retinoblastoma 1 (Rb1) gene plays an important role in fat development and adipogenesis in mice. Here, we cloned the partial cDNA sequences of the porcine Rb1 gene which contains the complete coding sequences (CDS) of 2820bp encoding a protein of 939 amino acids. Bioinformatic analysis revealed that the CDS of porcine Rb1 was highly identical with those of cattle, human and mice. The porcine Rb1 has three typical conserved structural domains, including Rb-A pocket domain, CYCLIN domain and C-terminus domain, and the phylogenetic tree indicates a closer genetic relationship with cattle and human. Tissue distribution analysis showed that Rb1 expression appeared to be ubiquitously in various tissues, being higher in heart, liver, muscle, and stomach. Furthermore, significant downregulation of Rb1 was found at the initial stage of dedifferentiated fat (DFAT) cells adipogenic differentiation. With the knockdown of the Rb1 expression by siRNA, the number of DFAT cells recruited to white rather than brown adipogenesis was promoted, and mRNA levels of adipogenic markers, such as PPARγ, aP2, LPL and adiponectin and protein expression of PPARγ and adiponectin were increased after hormone stimulation. The underlying mechanisms may be that knockdown of Rb1 promotes the mitotic clonal expansion and PPARγ expression by derepressing the transcriptional activity of E2F so as to facilitate the first steps of adipogenesis. In summary, we cloned and characterized an important negative regulator in adipogenic commitment of porcine DFAT cells.
Collapse
Affiliation(s)
- Xiaoming Hu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, People's Republic of China
| | - Pei Luo
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Xuewu Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Tongxing Song
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Yuanfei Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, People's Republic of China.
| | - Siwen Jiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; Key Laboratory of Swine Genetics and Breeding of Agricultural Ministry, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, People's Republic of China.
| |
Collapse
|
775
|
Resveratrol induces brown-like adipocyte formation in white fat through activation of AMP-activated protein kinase (AMPK) α1. Int J Obes (Lond) 2015; 39:967-76. [PMID: 25761413 PMCID: PMC4575949 DOI: 10.1038/ijo.2015.23] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/25/2014] [Accepted: 12/02/2014] [Indexed: 12/12/2022]
Abstract
Objective Development of brown-like/beige adipocytes in white adipose tissue (WAT) helps to reduce obesity. Thus, we investigated the effects of resveratrol, a dietary polyphenol capable of preventing obesity and related complications in humans and animal models, on brown-like adipocyte formation in inguinal WAT (iWAT). Methods CD1 female mice (5-month-old) were fed a high-fat diet with/without 0.1% resveratrol. In addition, primary stromal vascular cells separated from iWAT were subjected to resveratrol treatment. Markers of brown-like (beige) adipogenesis were measured and the involvement of AMP-activated protein kinase (AMPK) α1 was assessed using conditional knockout. Results Resveratrol significantly increased mRNA and/or protein expression of brown adipocyte markers including uncoupling protein 1 (UCP1), PR domain-containing 16 (PRDM16), Cell death-inducing DFFA-like effector A (Cidea), elongation of very long chain fatty acids protein 3 (Elovl3), peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), cytochrome C and pyruvate dehydrogenase (PDH) in differentiated iWAT stromal vascular cells (SVC), suggesting that resveratrol induced brown-like adipocyte formation in vitro. Concomitantly, resveratrol markedly enhanced AMPKα1 phosphorylation and differentiated SVC oxygen consumption. Such changes were absent in cells lacking AMPKα1, showing that AMPKα1 is a critical mediator of resveratrol action. Resveratrol also induced beige adipogenesis in vivo along with the appearance of multiocular adipocytes, increased UCP1 expression and enhanced fatty acid oxidation. Conclusion Resveratrol induces brown-like adipocyte formation in iWAT via AMPKα1 activation and suggest that its beneficial anti-obesity effects may be partly due to the browning of WAT and as a consequence, increased oxygen consumption.
Collapse
|
776
|
Berbée JFP, Boon MR, Khedoe PPSJ, Bartelt A, Schlein C, Worthmann A, Kooijman S, Hoeke G, Mol IM, John C, Jung C, Vazirpanah N, Brouwers LPJ, Gordts PLSM, Esko JD, Hiemstra PS, Havekes LM, Scheja L, Heeren J, Rensen PCN. Brown fat activation reduces hypercholesterolaemia and protects from atherosclerosis development. Nat Commun 2015; 6:6356. [PMID: 25754609 PMCID: PMC4366535 DOI: 10.1038/ncomms7356] [Citation(s) in RCA: 322] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/22/2015] [Indexed: 01/17/2023] Open
Abstract
Brown adipose tissue (BAT) combusts high amounts of fatty acids, thereby lowering plasma triglyceride levels and reducing obesity. However, the precise role of BAT in plasma cholesterol metabolism and atherosclerosis development remains unclear. Here we show that BAT activation by β3-adrenergic receptor stimulation protects from atherosclerosis in hyperlipidemic APOE*3-Leiden.CETP mice, a well-established model for human-like lipoprotein metabolism that unlike hyperlipidemic Apoe−/− and Ldlr−/− mice expresses functional apoE and LDLR. BAT activation increases energy expenditure and decreases plasma triglyceride and cholesterol levels. Mechanistically, we demonstrate that BAT activation enhances the selective uptake of fatty acids from triglyceride-rich lipoproteins into BAT, subsequently accelerating the hepatic clearance of the cholesterol-enriched remnants. These effects depend on a functional hepatic apoE-LDLR clearance pathway as BAT activation in Apoe−/− and Ldlr−/− mice does not attenuate hypercholesterolaemia and atherosclerosis. We conclude that activation of BAT is a powerful therapeutic avenue to ameliorate hyperlipidaemia and protect from atherosclerosis. Brown adipose tissue (BAT) produces heat by burning lipid triglycerides. Here, Berbée et al. show that pharmacological BAT activation protects hyperlipidemic mice from atherosclerosis, provided mice retain the metabolic capacity to clear cholesterol-enriched lipoprotein remnants by the liver.
Collapse
Affiliation(s)
- Jimmy F P Berbée
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Mariëtte R Boon
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - P Padmini S J Khedoe
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [3] Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Alexander Bartelt
- 1] Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany [2] Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Christian Schlein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany
| | - Sander Kooijman
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Geerte Hoeke
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Isabel M Mol
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Clara John
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany
| | - Caroline Jung
- Diagnostic and Interventional Radiology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany
| | - Nadia Vazirpanah
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Linda P J Brouwers
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Philip L S M Gordts
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, California 92093, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, California 92093, USA
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| | - Louis M Havekes
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [3] Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [4] Netherlands Organization for Applied Scientific Research-Metabolic Health Research, Gaubius Laboratory, Zernikedreef 9, Leiden 2333 CK, The Netherlands
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany
| | - Patrick C N Rensen
- 1] Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands [2] Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, Leiden 2333 ZA, The Netherlands
| |
Collapse
|
777
|
Ruan HB, Dietrich MO, Liu ZW, Zimmer MR, Li MD, Singh JP, Zhang K, Yin R, Wu J, Horvath TL, Yang X. O-GlcNAc transferase enables AgRP neurons to suppress browning of white fat. Cell 2015; 159:306-17. [PMID: 25303527 DOI: 10.1016/j.cell.2014.09.010] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 07/12/2014] [Accepted: 09/02/2014] [Indexed: 01/07/2023]
Abstract
Induction of beige cells causes the browning of white fat and improves energy metabolism. However, the central mechanism that controls adipose tissue browning and its physiological relevance are largely unknown. Here, we demonstrate that fasting and chemical-genetic activation of orexigenic AgRP neurons in the hypothalamus suppress the browning of white fat. O-linked β-N-acetylglucosamine (O-GlcNAc) modification of cytoplasmic and nuclear proteins regulates fundamental cellular processes. The levels of O-GlcNAc transferase (OGT) and O-GlcNAc modification are enriched in AgRP neurons and are elevated by fasting. Genetic ablation of OGT in AgRP neurons inhibits neuronal excitability through the voltage-dependent potassium channel, promotes white adipose tissue browning, and protects mice against diet-induced obesity and insulin resistance. These data reveal adipose tissue browning as a highly dynamic physiological process under central control, in which O-GlcNAc signaling in AgRP neurons is essential for suppressing thermogenesis to conserve energy in response to fasting.
Collapse
Affiliation(s)
- Hai-Bin Ruan
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Marcelo O Dietrich
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neurobiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 93042, Brazil
| | - Zhong-Wu Liu
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Marcelo R Zimmer
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 93042, Brazil
| | - Min-Dian Li
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Jay Prakash Singh
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Kaisi Zhang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Ruonan Yin
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Jing Wu
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neurobiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Xiaoyong Yang
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
778
|
Abstract
It was previously assumed that brown adipose tissue (BAT) is present in humans only for a short period following birth, the time in which mechanisms of generating heat by way of shivering are not yet developed. Although BAT is maximally recruited in early infancy, findings in recent years have led to a new consensus that metabolically active BAT remains present in most children and many adult humans. Evidence to date supports a slow and steady decline in BAT activity throughout life, with the exception of an intriguing spike in the prevalence and volume of BAT around the time of puberty that remains poorly understood. Because BAT activity is more commonly observed in individuals with a lower body mass index, an association seen in both adult and pediatric populations, there is the exciting possibility that BAT is protective against childhood and adult obesity. Indeed, the function and metabolic relevance of human BAT is currently an area of vigorous research. The goal of this review is to summarize what is currently known about changes that occur in BAT during various stages of life, with a particular emphasis on puberty and aging.
Collapse
Affiliation(s)
- Nicole H Rogers
- California Institute for Biomedical Research (Calibr) , La Jolla, CA 92037 , USA
| |
Collapse
|
779
|
Kong X, Yu J, Bi J, Qi H, Di W, Wu L, Wang L, Zha J, Lv S, Zhang F, Li Y, Hu F, Liu F, Zhou H, Liu J, Ding G. Glucocorticoids transcriptionally regulate miR-27b expression promoting body fat accumulation via suppressing the browning of white adipose tissue. Diabetes 2015; 64:393-404. [PMID: 25187367 PMCID: PMC4876791 DOI: 10.2337/db14-0395] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Long-term glucocorticoid (GC) treatment induces central fat accumulation and metabolic dysfunction. We demonstrate that microRNA-27b (miR-27b) plays a central role in the pathogenesis of GC-induced central fat accumulation. Overexpression of miR-27b had the same effects as dexamethasone (DEX) treatment on the inhibition of brown adipose differentiation and the energy expenditure of primary adipocytes. Conversely, antagonizing miR-27b function prevented DEX suppression of the expression of brown adipose tissue-specific genes. GCs transcriptionally regulate miR-27b expression through a GC receptor-mediated direct DNA-binding mechanism, and miR-27b suppresses browning of white adipose tissue (WAT) by targeting the three prime untranslated region of Prdm16. In vivo, antagonizing miR-27b function in DEX-treated mice resulted in the efficient induction of brown adipocytes within WAT and improved GC-induced central fat accumulation. Collectively, these results indicate that miR-27b functions as a central target of GC and as an upstream regulator of Prdm16 to control browning of WAT and, consequently, may represent a potential target in preventing obesity.
Collapse
Affiliation(s)
- Xiaocen Kong
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Jing Yu
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Jianhua Bi
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Hanmei Qi
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Wenjuan Di
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Lin Wu
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Long Wang
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Juanmin Zha
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Shan Lv
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Feng Zhang
- Department of General Surgery, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Yan Li
- Metabolic Syndrome Research Center of Central South University, Institute of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Fang Hu
- Metabolic Syndrome Research Center of Central South University, Institute of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Feng Liu
- Metabolic Syndrome Research Center of Central South University, Institute of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Juan Liu
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| | - Guoxian Ding
- Department of Geratology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
780
|
Peyrou M, Bourgoin L, Poher AL, Altirriba J, Maeder C, Caillon A, Fournier M, Montet X, Rohner-Jeanrenaud F, Foti M. Hepatic PTEN deficiency improves muscle insulin sensitivity and decreases adiposity in mice. J Hepatol 2015; 62:421-9. [PMID: 25234947 DOI: 10.1016/j.jhep.2014.09.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 09/03/2014] [Accepted: 09/09/2014] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS PTEN is a dual lipid/protein phosphatase, downregulated in steatotic livers with obesity or HCV infection. Liver-specific PTEN knockout (LPTEN KO) mice develop steatosis, inflammation/fibrosis and hepatocellular carcinoma with aging, but surprisingly also enhanced glucose tolerance. This study aimed at understanding the mechanisms by which hepatic PTEN deficiency improves glucose tolerance, while promoting fatty liver diseases. METHODS Control and LPTEN KO mice underwent glucose/pyruvate tolerance tests and euglycemic-hyperinsulinemic clamps. Body fat distribution was assessed by EchoMRI, CT-scan and dissection analyses. Primary/cultured hepatocytes and insulin-sensitive tissues were analysed ex vivo. RESULTS PTEN deficiency in hepatocytes led to steatosis through increased fatty acid (FA) uptake and de novo lipogenesis. Although LPTEN KO mice exhibited hepatic steatosis, they displayed increased skeletal muscle insulin sensitivity and glucose uptake, as assessed by euglycemic-hyperinsulinemic clamps. Surprisingly, white adipose tissue (WAT) depots were also drastically reduced. Analyses of key enzymes involved in lipid metabolism further indicated that FA synthesis/esterification was decreased in WAT. In addition, Ucp1 expression and multilocular lipid droplet structures were observed in this tissue, indicating the presence of beige adipocytes. Consistent with a liver to muscle/adipocyte crosstalk, the expression of liver-derived circulating factors, known to impact on muscle insulin sensitivity and WAT homeostasis (e.g. FGF21), was modulated in LPTEN KO mice. CONCLUSIONS Although steatosis develops in LPTEN KO mice, PTEN deficiency in hepatocytes promotes a crosstalk between liver and muscle, as well as adipose tissue, resulting in enhanced insulin sensitivity, improved glucose tolerance and decreased adiposity.
Collapse
Affiliation(s)
- Marion Peyrou
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Switzerland
| | - Lucie Bourgoin
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Switzerland
| | - Anne-Laure Poher
- Department of Internal Medicine Specialties, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Switzerland
| | - Jordi Altirriba
- Department of Internal Medicine Specialties, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Switzerland
| | - Christine Maeder
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Switzerland
| | - Aurélie Caillon
- Department of Internal Medicine Specialties, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Switzerland
| | - Margot Fournier
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Switzerland
| | - Xavier Montet
- Department of Radiology, Faculty of Medicine, University of Geneva, Switzerland
| | - Françoise Rohner-Jeanrenaud
- Department of Internal Medicine Specialties, Division of Endocrinology, Diabetology, Hypertension and Nutrition, Faculty of Medicine, University of Geneva, Switzerland
| | - Michelangelo Foti
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Switzerland.
| |
Collapse
|
781
|
The multifaceted factor peroxisome proliferator-activated receptor γ (PPARγ) in metabolism, immunity, and cancer. Arch Pharm Res 2015; 38:302-12. [DOI: 10.1007/s12272-015-0559-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 01/08/2015] [Indexed: 10/24/2022]
|
782
|
Papapetropoulos A, Hobbs AJ, Topouzis S. Extending the translational potential of targeting NO/cGMP-regulated pathways in the CVS. Br J Pharmacol 2015; 172:1397-414. [PMID: 25302549 DOI: 10.1111/bph.12980] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 09/08/2014] [Accepted: 10/05/2014] [Indexed: 02/06/2023] Open
Abstract
The discovery of NO as both an endogenous signalling molecule and as a mediator of the cardiovascular effects of organic nitrates was acknowledged in 1998 by the Nobel Prize in Physiology/Medicine. The characterization of its downstream signalling, mediated through stimulation of soluble GC (sGC) and cGMP generation, initiated significant translational interest, but until recently this was almost exclusively embodied by the use of PDE5 inhibitors in erectile dysfunction. Since then, research progress in two areas has contributed to an impressive expansion of the therapeutic targeting of the NO-sGC-cGMP axis: first, an increased understanding of the molecular events operating within this complex pathway and second, a better insight into its dys-regulation and uncoupling in human disease. Already-approved PDE5 inhibitors and novel, first-in-class molecules, which up-regulate the activity of sGC independently of NO and/or of the enzyme's haem prosthetic group, are undergoing clinical evaluation to treat pulmonary hypertension and myocardial failure. These molecules, as well as combinations or second-generation compounds, are also being assessed in additional experimental disease models and in patients in a wide spectrum of novel indications, such as endotoxic shock, diabetic cardiomyopathy and Becker's muscular dystrophy. There is well-founded optimism that the modulation of the NO-sGC-cGMP pathway will sustain the development of an increasing number of successful clinical candidates for years to come.
Collapse
|
783
|
Jeanson Y, Carrière A, Casteilla L. A New Role for Browning as a Redox and Stress Adaptive Mechanism? Front Endocrinol (Lausanne) 2015; 6:158. [PMID: 26500607 PMCID: PMC4598589 DOI: 10.3389/fendo.2015.00158] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/24/2015] [Indexed: 01/27/2023] Open
Abstract
The worldwide epidemic of obesity and metabolic disorders is focusing the attention of the scientific community on white adipose tissue (WAT) and its biology. This tissue is characterized not only by its capability to change in size and shape but also by its heterogeneity and versatility. WAT can be converted into brown fat-like tissue according to different physiological and pathophysiological situations. The expression of uncoupling protein-1 in brown-like adipocytes changes their function from energy storage to energy dissipation. This plasticity, named browning, was recently rediscovered and convergent recent accounts, including in humans, have revived the idea of using these oxidative cells to fight against metabolic diseases. Furthermore, recent reports suggest that, beside the increased energy dissipation and thermogenesis that may have adverse effects in situations such as cancer-associated cachexia and massive burns, browning could be also considered as an adaptive stress response to high redox pressure and to major stress that could help to maintain tissue homeostasis and integrity. The aim of this review is to summarize the current knowledge concerning brown adipocytes and the browning process and also to explore unexpected putative role(s) for these cells. While it is important to find new browning inducers to limit energy stores and metabolic diseases, it also appears crucial to develop new browning inhibitors to limit adverse energy dissipation in wasting-associated syndromes.
Collapse
Affiliation(s)
- Yannick Jeanson
- UMR STROMALab, CNRS 5273, INSERM U1031, Université Toulouse III – Paul Sabatier, Toulouse, France
| | - Audrey Carrière
- UMR STROMALab, CNRS 5273, INSERM U1031, Université Toulouse III – Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- UMR STROMALab, CNRS 5273, INSERM U1031, Université Toulouse III – Paul Sabatier, Toulouse, France
- *Correspondence: Louis Casteilla,
| |
Collapse
|
784
|
Varlamov O, Chu M, Cornea A, Sampath H, Roberts CT. Cell-autonomous heterogeneity of nutrient uptake in white adipose tissue of rhesus macaques. Endocrinology 2015; 156:80-9. [PMID: 25356825 PMCID: PMC4272393 DOI: 10.1210/en.2014-1699] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Phenotypic diversity may play an adaptive role by providing graded biological responses to fluctuations in environmental stimuli. We used single-cell imaging of the metabolizable fluorescent fatty acid analog 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY)-C12 and fluorescent 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-D-glucose (2-NBDG) to explore cellular heterogeneity in nutrient uptake in white adipose tissue (WAT) explants of rhesus macaques. Surprisingly, WAT displayed a striking cell size-independent mosaic pattern, in that adjacent adipocytes varied with respect to insulin-stimulated BODIPY-C12 and 2-NBDG uptake. Relative free fatty acid (FFA) transport activity correlated with the cellular levels of FFA transporter protein-1 and the scavenger receptor CD36 in individual adipocytes. In vitro incubation of WAT explants for 24 hours caused partial desynchronization of cellular responses, suggesting that adipocytes may slowly alter their differential nutrient uptake activity. In vitro-differentiated human adipocytes also exhibited a mosaic pattern of BODIPY-C12 uptake. WAT from animals containing a homogeneous population of large adipocytes was nonmosaic, in that every adipocyte exhibited a similar level of BODIPY-C12 fluorescence, suggesting that the development of obesity is associated with the loss of heterogeneity in WAT. Hence, for the first time, we demonstrate an intrinsic heterogeneity in FFA and glucose transport activity in WAT.
Collapse
Affiliation(s)
- Oleg Varlamov
- Divisions of Diabetes, Obesity, and Metabolism and Developmental and Reproductive Science (O.V., C.T.R.), and Division of Neuroscience (A.C.), Oregon National Primate Research Center, Beaverton, Oregon 97006; and Division of Endocrinology, Diabetes, and Clinical Nutrition, Department of Medicine (M.C., C.T.R.) and Center for Research Occupational and Environmental Toxicology (H.S.), Oregon Health and Science University, Portland, Oregon 97239
| | | | | | | | | |
Collapse
|
785
|
Peng XR, Gennemark P, O’Mahony G, Bartesaghi S. Unlock the Thermogenic Potential of Adipose Tissue: Pharmacological Modulation and Implications for Treatment of Diabetes and Obesity. Front Endocrinol (Lausanne) 2015; 6:174. [PMID: 26635723 PMCID: PMC4657528 DOI: 10.3389/fendo.2015.00174] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022] Open
Abstract
Brown adipose tissue (BAT) is considered an interesting target organ for the treatment of metabolic disease due to its high metabolic capacity. Non-shivering thermogenesis, once activated, can lead to enhanced partitioning and oxidation of fuels in adipose tissues, and reduce the burden of glucose and lipids on other metabolic organs such as liver, pancreas, and skeletal muscle. Sustained long-term activation of BAT may also lead to meaningful bodyweight loss. In this review, we discuss three different drug classes [the thiazolidinedione (TZD) class of PPARγ agonists, β3-adrenergic receptor agonists, and fibroblast growth factor 21 (FGF21) analogs] that have been proposed to regulate BAT and beige recruitment or activation, or both, and which have been tested in both rodent and human. The learnings from these classes suggest that restoration of functional BAT and beige mass as well as improved activation might be required to fully realize the metabolic potential of these tissues. Whether this can be achieved without the undesired cardiovascular side effects exhibited by the TZD PPARγ agonists and β3-adrenergic receptor agonists remains to be resolved.
Collapse
Affiliation(s)
- Xiao-Rong Peng
- Cardiovascular and Metabolic Diseases IMED Biotech Unit, Diabetes Bioscience Department, AstraZeneca R&D, Mölndal, Sweden
- *Correspondence: Xiao-Rong Peng,
| | - Peter Gennemark
- Cardiovascular and Metabolic Diseases IMED Biotech Unit, Drug Metabolism and Pharmacokinetics Department, AstraZeneca R&D, Mölndal, Sweden
| | - Gavin O’Mahony
- Cardiovascular and Metabolic Diseases IMED Biotech Unit, Medicinal Chemistry Department, AstraZeneca R&D, Mölndal, Sweden
| | - Stefano Bartesaghi
- Cardiovascular and Metabolic Diseases IMED Biotech Unit, Diabetes Bioscience Department, AstraZeneca R&D, Mölndal, Sweden
| |
Collapse
|
786
|
Fernández-Galilea M, Pérez-Matute P, Prieto-Hontoria PL, Houssier M, Burrell MA, Langin D, Martínez JA, Moreno-Aliaga MJ. α-Lipoic acid treatment increases mitochondrial biogenesis and promotes beige adipose features in subcutaneous adipocytes from overweight/obese subjects. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:273-81. [PMID: 25542506 DOI: 10.1016/j.bbalip.2014.12.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/10/2014] [Accepted: 12/15/2014] [Indexed: 01/20/2023]
Abstract
α-Lipoic acid (α-Lip) is a natural occurring antioxidant with beneficial anti-obesity properties. The aim of this study was to investigate the putative effects of α-Lip on mitochondrial biogenesis and the acquirement of brown-like characteristics by subcutaneous adipocytes from overweight/obese subjects. Thus, fully differentiated human subcutaneous adipocytes were treated with α-Lip (100 and 250μM) for 24h for studies on mitochondrial content and morphology, mitochondrial DNA (mtDNA) copy number, fatty acid oxidation enzymes and brown/beige characteristic genes. The involvement of the Sirtuin1/Peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (SIRT1/PGC-1α) pathway was also evaluated. Our results showed that α-Lip increased mitochondrial content in cultured human adipocytes as revealed by electron microscopy and by mitotracker green labeling. Moreover, an enhancement in mtDNA content was observed. This increase was accompanied by an up-regulation of SIRT1 protein levels, a decrease in PGC-1α acetylation and up-regulation of Nuclear respiratory factor 1 (Nrf1) and Mitochondrial transcription factor (Tfam) transcription factors. Enhanced oxygen consumption and fatty acid oxidation enzymes, Carnitine palmitoyl transferase 1 and Acyl-coenzyme A oxidase (CPT-1 and ACOX) were also observed. Mitochondria from α-Lip-treated adipocytes exhibited some morphological characteristics of brown mitochondria, and α-Lip also induced up-regulation of some brown/beige adipocytes markers such as cell death-inducing DFFA-like effector a (Cidea) and T-box 1 (Tbx1). Moreover, α-Lip up-regulated PR domain containing 16 (Prdm16) mRNA levels in treated adipocytes. Therefore, our study suggests the ability of α-Lip to promote mitochondrial biogenesis and brown-like remodeling in cultured white subcutaneous adipocytes from overweight/obese donors.
Collapse
Affiliation(s)
- Marta Fernández-Galilea
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain
| | - Patricia Pérez-Matute
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Pedro L Prieto-Hontoria
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Faculty of Health and Physical Activity Science, University SEK, Santiago, Chile
| | - Marianne Houssier
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | - María A Burrell
- Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | - Dominique Langin
- INSERM, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France; Toulouse University Hospitals, Laboratory of Clinical Biochemistry, Toulouse, France
| | - J Alfredo Martínez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Center for Nutrition Research, University of Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Center for Nutrition Research, University of Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
787
|
Kliewer KL, Ke JY, Tian M, Cole RM, Andridge RR, Belury MA. Adipose tissue lipolysis and energy metabolism in early cancer cachexia in mice. Cancer Biol Ther 2014; 16:886-97. [PMID: 25457061 DOI: 10.4161/15384047.2014.987075] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cancer cachexia is a progressive metabolic disorder that results in depletion of adipose tissue and skeletal muscle. A growing body of literature suggests that maintaining adipose tissue mass in cachexia may improve quality-of-life and survival outcomes. Studies of lipid metabolism in cachexia, however, have generally focused on later stages of the disorder when severe loss of adipose tissue has already occurred. Here, we investigated lipid metabolism in adipose, liver and muscle tissues during early stage cachexia - before severe fat loss - in the colon-26 murine model of cachexia. White adipose tissue mass in cachectic mice was moderately reduced (34-42%) and weight loss was less than 10% of initial body weight in this study of early cachexia. In white adipose depots of cachectic mice, we found evidence of enhanced protein kinase A - activated lipolysis which coincided with elevated total energy expenditure and increased expression of markers of brown (but not white) adipose tissue thermogenesis and the acute phase response. Total lipids in liver and muscle were unchanged in early cachexia while markers of fatty oxidation were increased. Many of these initial metabolic responses contrast with reports of lipid metabolism in later stages of cachexia. Our observations suggest intervention studies to preserve fat mass in cachexia should be tailored to the stage of cachexia. Our observations also highlight a need for studies that delineate the contribution of cachexia stage and animal model to altered lipid metabolism in cancer cachexia and identify those that most closely mimic the human condition.
Collapse
Key Words
- ACOX, acyl CoA oxidase
- ATGL, adipose triglyceride lipase
- COX, cytochrome c oxidase subunits
- CPT, carnitine palmitoyltransferase
- CRP, C-reactive protein
- DIO, iodothyronine deiodinase
- GYK, glycerokinase
- H&E, hematoxylin and eosin
- HSL, hormone sensitive lipase
- LPL, lipoprotein lipase
- MuRF, muscle ring finger protein
- PGC, peroxisome proliferator activated receptor gamma coactivator
- PKA, protein kinase A
- PPAR, peroxisome proliferator activated receptor
- PRDM, PR domain zinc finger protein
- RER, respiratory exchange ratio.
- TEE, total energy expenditure
- UCP, uncoupling protein
- colon-26 adenocarcinoma
- eWAT, epididymal white adipose tissue
- early cachexia
- energy expenditure
- iBAT, interscapular brown adipose tissue
- iWAT, inguinal white adipose tissue
- lipid metabolism
- lipolysis
- thermogenesis
Collapse
Affiliation(s)
- Kara L Kliewer
- a Department of Human Sciences ; College of Education and Human Ecology ; The Ohio State University ; Columbus , OH USA
| | | | | | | | | | | |
Collapse
|
788
|
Wang Y, Zhu T, Ke S, Fang N, Irwin DM, Lei M, Zhang J, Shi H, Zhang S, Wang Z. The great roundleaf bat (Hipposideros armiger) as a good model for cold-induced browning of intra-abdominal white adipose tissue. PLoS One 2014; 9:e112495. [PMID: 25393240 PMCID: PMC4231071 DOI: 10.1371/journal.pone.0112495] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Inducing beige fat from white adipose tissue (WAT) is considered to be a shortcut to weight loss and increasingly becoming a key area in research into treatments for obesity and related diseases. However, currently, animal models of beige fat are restricted to rodents, where subcutaneous adipose tissue (sWAT, benign WAT) is more liable to develop into the beige fat under specific activators than the intra-abdominal adipose tissue (aWAT, malignant WAT) that is the major source of obesity related diseases in humans. METHODS Here we induced beige fat by cold exposure in two species of bats, the great roundleaf bat (Hipposideros armiger) and the rickett's big-footed bat (Myotis ricketti), and compared the molecular and morphological changes with those seen in the mouse. Expression of thermogenic genes (Ucp1 and Pgc1a) was measured by RT-qPCR and adipocyte morphology examined by HE staining at three adipose locations, sWAT, aWAT and iBAT (interscapular brown adipose tissue). RESULTS Expression of Ucp1 and Pgc1a was significantly upregulated, by 729 and 23 fold, respectively, in aWAT of the great roundleaf bat after exposure to 10°C for 7 days. Adipocyte diameters of WATs became significantly reduced and the white adipocytes became brown-like in morphology. In mice, similar changes were found in the sWAT, but much lower amounts of changes in aWAT were seen. Interestingly, the rickett's big-footed bat did not show such a tendency in beige fat. CONCLUSIONS The great roundleaf bat is potentially a good animal model for human aWAT browning research. Combined with rodent models, this model should be helpful for finding therapies for reducing harmful aWAT in humans.
Collapse
Affiliation(s)
- Yao Wang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Tengteng Zhu
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Shanshan Ke
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Na Fang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Ming Lei
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Junpeng Zhang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Huizhen Shi
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
| | - Shuyi Zhang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
- * E-mail: (SZ); (ZW)
| | - Zhe Wang
- Institute of Molecular Ecology and Evolution, East China Normal University, Shanghai, China
- * E-mail: (SZ); (ZW)
| |
Collapse
|
789
|
Aromatase controls Sjögren syndrome-like lesions through monocyte chemotactic protein-1 in target organ and adipose tissue-associated macrophages. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:151-61. [PMID: 25447050 DOI: 10.1016/j.ajpath.2014.09.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 09/02/2014] [Accepted: 09/09/2014] [Indexed: 02/06/2023]
Abstract
Several autoimmune diseases are known to develop in postmenopausal women. However, the mechanism by which estrogen deficiency influences autoimmunity is unknown. Aromatase is an enzyme that converts androgens to estrogens. Herein, we used female aromatase gene knockout (ArKO) mice as a model of estrogen deficiency to investigate the molecular mechanism that underlies the onset and development of autoimmunity. Histological analyses showed that inflammatory lesions in the lacrimal and salivary glands of ArKO mice increased with age. Adoptive transfer of spleen cells or bone marrow cells from ArKO mice into recombination activating gene 2 knockout mice failed to induce the autoimmune lesions. Expression of mRNA encoding proinflammatory cytokines and monocyte chemotactic protein-1 increased in white adipose tissue of ArKO mice and was significantly higher than that in wild-type mice. Moreover, an increased number of inflammatory M1 macrophages was observed in white adipose tissue of ArKO mice. A significantly increased monocyte chemotactic protein-1 mRNA expression of the salivary gland tissue in ArKO was found together with adiposity. Furthermore, the autoimmune lesions in a murine model of Sjögren syndrome were exacerbated by administration of an aromatase inhibitor. These results suggest that aromatase may play a key role in the pathogenesis of Sjögren syndrome-like lesions by controlling the target organ and adipose tissue-associated macrophage.
Collapse
|
790
|
Gnad T, Scheibler S, von Kügelgen I, Scheele C, Kilić A, Glöde A, Hoffmann LS, Reverte-Salisa L, Horn P, Mutlu S, El-Tayeb A, Kranz M, Deuther-Conrad W, Brust P, Lidell ME, Betz MJ, Enerbäck S, Schrader J, Yegutkin GG, Müller CE, Pfeifer A. Adenosine activates brown adipose tissue and recruits beige adipocytes via A2A receptors. Nature 2014; 516:395-9. [DOI: 10.1038/nature13816] [Citation(s) in RCA: 263] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023]
|
791
|
Fromenty B. Burn after feeding. An old uncoupler of oxidative phosphorylation is redesigned for the treatment of nonalcoholic fatty liver disease. Clin Res Hepatol Gastroenterol 2014; 38:545-9. [PMID: 24924900 DOI: 10.1016/j.clinre.2014.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/17/2014] [Accepted: 04/28/2014] [Indexed: 02/04/2023]
Abstract
Uncoupling of oxidative phosphorylation (OXPHOS) in brown adipose tissue can be used by hibernating animals to produce heat at the expense of their fat mass. In a recent work, Dr Shulman et al. generated a liver-targeted derivative of the prototypical OXPHOS uncoupler 2,4-dinitrophenol that alleviated steatosis, hypertriglyceridemia and insulin resistance in several models of nonalcoholic fatty liver disease and type 2 diabetes.
Collapse
Affiliation(s)
- B Fromenty
- Inserm, U991, université de Rennes 1, 35000 Rennes, France.
| |
Collapse
|
792
|
Abstract
A detailed appreciation of the control of adipose tissue whether it be white, brown or brite/beige has never been more important to the development of a framework on which to build therapeutic strategies to combat obesity. This is because 1) the rate of fatty acid release into the circulation from lipolysis in white adipose tissue (WAT) is integrally important to the development of obesity, 2) brown adipose tissue (BAT) has now moved back to center stage with the realization that it is present in adult humans and, in its activated form, is inversely proportional to levels of obesity and 3) the identification and characterization of "brown-like" or brite/beige fat is likely to be one of the most exciting developments in adipose tissue biology in the last decade. Central to all of these developments is the role of the CNS in the control of different fat cell functions and central to CNS control is the integrative capacity of the hypothalamus. In this chapter we will attempt to detail key issues relevant to the structure and function of hypothalamic and downstream control of WAT and BAT and highlight the importance of developing an understanding of the neural input to brite/beige fat cells as a precursor to its recruitment as therapeutic target.
Collapse
Affiliation(s)
- A Stefanidis
- Department of Physiology, Monash University, Clayton, 3800, Australia
| | - N M Wiedmann
- Department of Physiology, Monash University, Clayton, 3800, Australia
| | - E S Adler
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - B J Oldfield
- Department of Physiology, Monash University, Clayton, 3800, Australia.
| |
Collapse
|
793
|
Yang X, Bi P, Kuang S. Fighting obesity: When muscle meets fat. Adipocyte 2014; 3:280-9. [PMID: 26317052 DOI: 10.4161/21623945.2014.964075] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/13/2014] [Accepted: 09/04/2014] [Indexed: 12/25/2022] Open
Abstract
The prevalence of obesity has risen to an unprecedented level. According to World Health Organization, over 500 million adults, equivalent to 10%-14% of the world population, were obese with a body mass index (BMI) of 30 kg/m(2) or greater in 2008.(1) This rising prevalence and earlier onset of obesity is believed to be resulted from an interplay of genetic factors, over-nutrition and physical inactivity in modern lifestyles. Obesity also increases the susceptibility to metabolic syndromes, hypertension, cardiovascular diseases, Type 2 diabetes mellitus (T2DM) and cancer.(2-4) The global obesity epidemic has sparked substantial interests in the biology of adipose tissue (fat). In addition, the skeletal muscle and its secretive factors (myokines) have also been shown to play a critical role in controlling body energy balance, adipose homeostasis and inflammation status.(5) Interestingly, skeletal muscle cells share a common developmental origin with brown adipocytes,(6,7) which breaks down lipids to generate heat - thus reducing obesity. Here, we provide a brief overview of the basics and recent progress in muscle-fat crosstalk in the context of body energy metabolism, obesity, and diabetes. We summarize the different types of adipocytes, their developmental origins and implications in body composition. We highlight the role of several novel myokines in regulating fat mass and systemic energy balance, and evaluate the potential of skeletal muscles as a therapeutic target to treat obesity.
Collapse
|
794
|
Carrière A, Jeanson Y, Berger-Müller S, André M, Chenouard V, Arnaud E, Barreau C, Walther R, Galinier A, Wdziekonski B, Villageois P, Louche K, Collas P, Moro C, Dani C, Villarroya F, Casteilla L. Browning of white adipose cells by intermediate metabolites: an adaptive mechanism to alleviate redox pressure. Diabetes 2014; 63:3253-65. [PMID: 24789919 DOI: 10.2337/db13-1885] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The presence of brown adipose tissue (BAT) in human adults opens attractive perspectives to treat metabolic disorders. Indeed, BAT dissipates energy as heat via uncoupling protein (UCP)1. Brown adipocytes are located in specific deposits or can emerge among white fat through the so-called browning process. Although numerous inducers have been shown to drive this process, no study has investigated whether it could be controlled by specific metabolites. Here, we show that lactate, an important metabolic intermediate, induces browning of murine white adipose cells with expression of functional UCP1. Lactate-induced browning also occurs in human cells and in vivo. Lactate controls Ucp1 expression independently of hypoxia-inducible factor-1α and PPARα pathways but requires active PPARγ signaling. We demonstrate that the lactate effect on Ucp1 is mediated by intracellular redox modifications as a result of lactate transport through monocarboxylate transporters. Further, the ketone body β-hydroxybutyrate, another metabolite that impacts redox state, is also a strong browning inducer. Because this redox-dependent increase in Ucp1 expression promotes an oxidative phenotype with mitochondria, browning appears as an adaptive mechanism to alleviate redox pressure. Our findings open new perspectives for the control of adipose tissue browning and its physiological relevance.
Collapse
Affiliation(s)
- Audrey Carrière
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Yannick Jeanson
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Sandra Berger-Müller
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Mireille André
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Vanessa Chenouard
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Emmanuelle Arnaud
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Corinne Barreau
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Romy Walther
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Anne Galinier
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| | - Brigitte Wdziekonski
- Faculté de Médecine, Institut de Biologie Valrose CNRS/INSERM/Université Nice Sophia Antipolis, Nice, France
| | - Phi Villageois
- Faculté de Médecine, Institut de Biologie Valrose CNRS/INSERM/Université Nice Sophia Antipolis, Nice, France
| | - Katie Louche
- INSERM, UMR 1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Philippe Collas
- Stem Cell Epigenetics Laboratory, Institute of Basic Medical Sciences, Faculty of Medicine, and Norwegian Center for Stem Cell Research, University of Oslo, Oslo, Norway
| | - Cédric Moro
- INSERM, UMR 1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France
| | - Christian Dani
- Faculté de Médecine, Institut de Biologie Valrose CNRS/INSERM/Université Nice Sophia Antipolis, Nice, France
| | - Francesc Villarroya
- Departament de Bioquimica i Biologia Molecular and Institute of Biomedicine, Universitat de Barcelona, and CIBER Fisiopatología de la Obesidad y Nutrición, Barcelona, Spain
| | - Louis Casteilla
- CNRS 5273, UMR STROMALab, Toulouse, France Université de Toulouse, Université Paul Sabatier, UMR 5273, Toulouse, France INSERM U1031, Toulouse, France Etablissement Français du Sang Pyrénées-Méditerranée, Toulouse, France
| |
Collapse
|
795
|
Leptin administration activates irisin-induced myogenesis via nitric oxide-dependent mechanisms, but reduces its effect on subcutaneous fat browning in mice. Int J Obes (Lond) 2014; 39:397-407. [DOI: 10.1038/ijo.2014.166] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 08/06/2014] [Accepted: 09/02/2014] [Indexed: 02/06/2023]
|
796
|
Heine M, Bartelt A, Bruns OT, Bargheer D, Giemsa A, Freund B, Scheja L, Waurisch C, Eychmüller A, Reimer R, Weller H, Nielsen P, Heeren J. The cell-type specific uptake of polymer-coated or micelle-embedded QDs and SPIOs does not provoke an acute pro-inflammatory response in the liver. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2014; 5:1432-1440. [PMID: 25247125 PMCID: PMC4168844 DOI: 10.3762/bjnano.5.155] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 08/12/2014] [Indexed: 05/27/2023]
Abstract
Semiconductor quantum dots (QD) and superparamagnetic iron oxide nanocrystals (SPIO) have exceptional physical properties that are well suited for biomedical applications in vitro and in vivo. For future applications, the direct injection of nanocrystals for imaging and therapy represents an important entry route into the human body. Therefore, it is crucial to investigate biological responses of the body to nanocrystals to avoid harmful side effects. In recent years, we established a system to embed nanocrystals with a hydrophobic oleic acid shell either by lipid micelles or by the amphiphilic polymer poly(maleic anhydride-alt-1-octadecene) (PMAOD). The goal of the current study is to investigate the uptake processes as well as pro-inflammatory responses in the liver after the injection of these encapsulated nanocrystals. By immunofluorescence and electron microscopy studies using wild type mice, we show that 30 min after injection polymer-coated nanocrystals are primarily taken up by liver sinusoidal endothelial cells. In contrast, by using wild type, Ldlr (-/-) as well as Apoe (-/-) mice we show that nanocrystals embedded within lipid micelles are internalized by Kupffer cells and, in a process that is dependent on the LDL receptor and apolipoprotein E, by hepatocytes. Gene expression analysis of pro-inflammatory markers such as tumor necrosis factor alpha (TNFα) or chemokine (C-X-C motif) ligand 10 (Cxcl10) indicated that 48 h after injection internalized nanocrystals did not provoke pro-inflammatory pathways. In conclusion, internalized nanocrystals at least in mouse liver cells, namely endothelial cells, Kupffer cells and hepatocytes are at least not acutely associated with potential adverse side effects, underlining their potential for biomedical applications.
Collapse
Affiliation(s)
- Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
| | - Alexander Bartelt
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
- Department of Genetics and Complex Disease, Harvard School of Public Health, 665 Huntington Avenue, Boston, 02115 MA, USA
| | - Oliver T Bruns
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | - Denise Bargheer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
| | - Artur Giemsa
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
| | - Barbara Freund
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
| | - Christian Waurisch
- Institute of Physical Chemistry and Electrochemistry, Technical University of Dresden, 01062 Dresden, Germany
| | - Alexander Eychmüller
- Institute of Physical Chemistry and Electrochemistry, Technical University of Dresden, 01062 Dresden, Germany
| | - Rudolph Reimer
- Department of Electron Microscopy and Micro Technology, Heinrich-Pette Institute, Martinistrasse 52, 20246 Hamburg, Germany
| | - Horst Weller
- Institute of Physical Chemistry, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany
| | - Peter Nielsen
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf; Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
797
|
Pfeifer A, Hoffmann LS. Brown, beige, and white: the new color code of fat and its pharmacological implications. Annu Rev Pharmacol Toxicol 2014; 55:207-27. [PMID: 25149919 DOI: 10.1146/annurev-pharmtox-010814-124346] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Brown adipose tissue (BAT) was previously regarded as a special type of fat relevant only for defending hibernating animals and newborns against a cold environment. Recently, BAT has received considerable attention following its (re)discovery in humans. Using glucose tracers, multiple laboratories independently found metabolically active BAT in adults. The enormous metabolic powers of BAT in animal models could make it an attractive target for antiobesity therapies in humans. Here, we review the present knowledge on the role of BAT in energy homeostasis and metabolism, focusing on signaling pathways and potential targets for novel therapeutics. We also shine light on ongoing debates, including those about the true color of brown fat in adults, as well as on the requirements for translation of basic research on BAT into clinical medicine.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, 53105 Bonn, Germany;
| | | |
Collapse
|
798
|
Li Y, Fromme T, Schweizer S, Schöttl T, Klingenspor M. Taking control over intracellular fatty acid levels is essential for the analysis of thermogenic function in cultured primary brown and brite/beige adipocytes. EMBO Rep 2014; 15:1069-76. [PMID: 25135951 DOI: 10.15252/embr.201438775] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Thermogenesis in brown adipocytes, conferred by mitochondrial uncoupling protein 1 (UCP1), is receiving great attention because metabolically active brown adipose tissue may protect humans from metabolic diseases. In particular, the thermogenic function of brown-like adipocytes in white adipose tissue, known as brite (or beige) adipocytes, is currently of prime interest. A valid procedure to quantify the specific contribution of UCP1 to thermogenesis is thus of vital importance. Adrenergic stimulation of lipolysis is a common way to activate UCP1. We here report, however, that in this frequently applied setup, taking control over intracellular fatty acid levels is essential for the analysis of thermogenic function in cultured brown and brite adipocytes. By the application of these findings, we demonstrate that UCP1 is functionally thermogenic in intact brite adipocytes and adrenergic UCP1 activation is largely dependent on adipose triglyceride lipase (ATGL) rather than hormone sensitive lipase (HSL).
Collapse
Affiliation(s)
- Yongguo Li
- Molecular Nutritional Medicine, Else Kröner Fresenius Center for Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Tobias Fromme
- Molecular Nutritional Medicine, Else Kröner Fresenius Center for Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Sabine Schweizer
- Molecular Nutritional Medicine, Else Kröner Fresenius Center for Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Theresa Schöttl
- Molecular Nutritional Medicine, Else Kröner Fresenius Center for Nutritional Medicine, Technische Universität München, Freising, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner Fresenius Center for Nutritional Medicine, Technische Universität München, Freising, Germany
| |
Collapse
|
799
|
Gunawardana SC. Benefits of healthy adipose tissue in the treatment of diabetes. World J Diabetes 2014; 5:420-430. [PMID: 25126390 PMCID: PMC4127579 DOI: 10.4239/wjd.v5.i4.420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/24/2014] [Accepted: 06/03/2014] [Indexed: 02/05/2023] Open
Abstract
The major malfunction in diabetes mellitus is severe perturbation of glucose homeostasis caused by deficiency of insulin. Insulin deficiency is either absolute due to destruction or failure of pancreatic β cells, or relative due to decreased sensitivity of peripheral tissues to insulin. The primary lesion being related to insulin, treatments for diabetes focus on insulin replacement and/or increasing sensitivity to insulin. These therapies have their own limitations and complications, some of which can be life-threatening. For example, exogenous insulin administration can lead to fatal hypoglycemic episodes; islet/pancreas transplantation requires life-long immunosuppressive therapy; and anti-diabetic drugs have dangerous side effects including edema, heart failure and lactic acidosis. Thus the need remains for better safer long term treatments for diabetes. The ultimate goal in treating diabetes is to re-establish glucose homeostasis, preferably through endogenously generated hormones. Recent studies increasingly show that extra-pancreatic hormones, particularly those arising from adipose tissue, can compensate for insulin, or entirely replace the function of insulin under appropriate circumstances. Adipose tissue is a versatile endocrine organ that secretes a variety of hormones with far-reaching effects on overall metabolism. While unhealthy adipose tissue can exacerbate diabetes through limiting circulation and secreting of pro-inflammatory cytokines, healthy uninflamed adipose tissue secretes beneficial adipokines with hypoglycemic and anti-inflammatory properties, which can complement and/or compensate for the function of insulin. Administration of specific adipokines is known to alleviate both type 1 and 2 diabetes, and leptin mono-therapy is reported to reverse type 1 diabetes independent of insulin. Although specific adipokines may correct diabetes, administration of individual adipokines still carries risks similar to those of insulin monotherapy. Thus a better approach is to achieve glucose homeostasis with endogenously-generated adipokines through transplantation or regeneration of healthy adipose tissue. Our recent studies on mouse models show that type 1 diabetes can be reversed without insulin through subcutaneous transplantation of embryonic brown adipose tissue, which leads to replenishment of recipients’ white adipose tissue; increase of a number of beneficial adipokines; and fast and long-lasting euglycemia. Insulin-independent glucose homeostasis is established through a combination of endogenously generated hormones arising from the transplant and/or newly-replenished white adipose tissue. Transplantation of healthy white adipose tissue is reported to alleviate type 2 diabetes in rodent models on several occasions, and increasing the content of endogenous brown adipose tissue is known to combat obesity and type 2 diabetes in both humans and animal models. While the underlying mechanisms are not fully documented, the beneficial effects of healthy adipose tissue in improving metabolism are increasingly reported, and are worthy of attention as a powerful tool in combating metabolic disease.
Collapse
|
800
|
Kim KH, Lee MS. FGF21 as a Stress Hormone: The Roles of FGF21 in Stress Adaptation and the Treatment of Metabolic Diseases. Diabetes Metab J 2014; 38:245-51. [PMID: 25215270 PMCID: PMC4160577 DOI: 10.4093/dmj.2014.38.4.245] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fibroblast growth factor 21 (FGF21) is an endocrine hormone that is primarily expressed in the liver and exerts beneficial effects on obesity and related metabolic diseases. In addition to its remarkable pharmacologic actions, the physiological roles of FGF21 include the maintenance of energy homeostasis in the body in conditions of metabolic or environmental stress. The expression of FGF21 is induced in multiple organs in response to diverse physiological or pathological stressors, such as starvation, nutrient excess, autophagy deficiency, mitochondrial stress, exercise, and cold exposure. Thus, the FGF21 induction caused by stress plays an important role in adaptive response to these stimuli. Here, we highlight our current understanding of the functional importance of the induction of FGF21 by diverse stressors as a feedback mechanism that prevents excessive stress.
Collapse
Affiliation(s)
- Kook Hwan Kim
- Department of Medicine, Samsung Medical Center and Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Shik Lee
- Department of Medicine, Samsung Medical Center and Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|