51
|
Oguri Y, Fujita Y, Abudukadier A, Ohashi A, Goto T, Furuya F, Obara A, Fukushima T, Matsuo N, Kim M, Hosokawa M, Kawada T, Hasegawa H, Inagaki N. Tetrahydrobiopterin activates brown adipose tissue and regulates systemic energy metabolism. JCI Insight 2017; 2:91981. [PMID: 28469071 PMCID: PMC5414566 DOI: 10.1172/jci.insight.91981] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/29/2017] [Indexed: 12/12/2022] Open
Abstract
Brown adipose tissue (BAT) is a central organ that acts to increase energy expenditure; its regulatory factors could be clinically useful in the treatment of obesity. Tetrahydrobiopterin (BH4) is an essential cofactor of tyrosine hydroxylase and nitric oxide synthase (NOS). Although BH4 regulates the known regulatory factors of BAT, such as noradrenaline (NA) and NO, participation of BH4 in BAT function remains unclear. In the present study, we investigate the role of BH4 in the regulation of BAT. Hph-1 mice, a mouse model of BH4 deficiency, exhibit obesity, adiposity, glucose intolerance, insulin resistance, and impaired BAT function. Impaired BAT function was ameliorated together with systemic metabolic disturbances by BAT transplantation from BH4-sufficient mice (control mice) into BH4-deficient mice, strongly suggesting that BH4-induced BAT has a critical role in the regulation of systemic energy metabolism. Both NA derived from the sympathetic nerve and NO derived from endothelial NOS in the blood vessels participate in the regulation of BH4. In addition, a direct effect of BH4 in the stimulation of brown adipocytes via NO is implicated. Taken together, BH4 activates BAT and regulates systemic energy metabolism; this suggests an approach for metabolic disorders, such as obesity and diabetes.
Collapse
Affiliation(s)
- Yasuo Oguri
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yoshihito Fujita
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Abulizi Abudukadier
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akiko Ohashi
- Department of Anatomy, Nihon University School of Dentistry, Tokyo, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Futoshi Furuya
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akio Obara
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toru Fukushima
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naomi Matsuo
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Minji Kim
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masaya Hosokawa
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Faculty of Human Sciences, Tezukayama Gakuin University, Osaka, Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Hiroyuki Hasegawa
- Department of Anatomy, Nihon University School of Dentistry, Tokyo, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
52
|
Sim CK, Kim SY, Brunmeir R, Zhang Q, Li H, Dharmasegaran D, Leong C, Lim YY, Han W, Xu F. Regulation of white and brown adipocyte differentiation by RhoGAP DLC1. PLoS One 2017; 12:e0174761. [PMID: 28358928 PMCID: PMC5373604 DOI: 10.1371/journal.pone.0174761] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/15/2017] [Indexed: 12/22/2022] Open
Abstract
Adipose tissues constitute an important component of metabolism, the dysfunction of which can cause obesity and type II diabetes. Here we show that differentiation of white and brown adipocytes requires Deleted in Liver Cancer 1 (DLC1), a Rho GTPase Activating Protein (RhoGAP) previously studied for its function in liver cancer. We identified Dlc1 as a super-enhancer associated gene in both white and brown adipocytes through analyzing the genome-wide binding profiles of PPARγ, the master regulator of adipogenesis. We further observed that Dlc1 expression increases during differentiation, and knockdown of Dlc1 by siRNA in white adipocytes reduces the formation of lipid droplets and the expression of fat marker genes. Moreover, knockdown of Dlc1 in brown adipocytes reduces expression of brown fat-specific genes and diminishes mitochondrial respiration. Dlc1-/- knockout mouse embryonic fibroblasts show a complete inability to differentiate into adipocytes, but this phenotype can be rescued by inhibitors of Rho-associated kinase (ROCK) and filamentous actin (F-actin), suggesting the involvement of Rho pathway in DLC1-regulated adipocyte differentiation. Furthermore, PPARγ binds to the promoter of Dlc1 gene to regulate its expression during both white and brown adipocyte differentiation. These results identify DLC1 as an activator of white and brown adipocyte differentiation, and provide a molecular link between PPARγ and Rho pathways.
Collapse
Affiliation(s)
- Choon Kiat Sim
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sun-Yee Kim
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A*STAR, Singapore
| | - Reinhard Brunmeir
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Qiongyi Zhang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Hongyu Li
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A*STAR, Singapore
| | - Dharmini Dharmasegaran
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Carol Leong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ying Yan Lim
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Weiping Han
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A*STAR, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Feng Xu
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- * E-mail:
| |
Collapse
|
53
|
Ziętak M, Chabowska-Kita A, Kozak LP. Brown fat thermogenesis: Stability of developmental programming and transient effects of temperature and gut microbiota in adults. Biochimie 2017; 134:93-98. [DOI: 10.1016/j.biochi.2016.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/09/2016] [Indexed: 12/23/2022]
|
54
|
Perdikari A, Kulenkampff E, Rudigier C, Neubauer H, Luippold G, Redemann N, Wolfrum C. A high-throughput, image-based screen to identify kinases involved in brown adipocyte development. Sci Signal 2017; 10:10/466/eaaf5357. [DOI: 10.1126/scisignal.aaf5357] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
55
|
Chung YW, Ahmad F, Tang Y, Hockman SC, Kee HJ, Berger K, Guirguis E, Choi YH, Schimel DM, Aponte AM, Park S, Degerman E, Manganiello VC. White to beige conversion in PDE3B KO adipose tissue through activation of AMPK signaling and mitochondrial function. Sci Rep 2017; 7:40445. [PMID: 28084425 PMCID: PMC5234021 DOI: 10.1038/srep40445] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/18/2016] [Indexed: 12/21/2022] Open
Abstract
Understanding mechanisms by which a population of beige adipocytes is increased in white adipose tissue (WAT) reflects a potential strategy in the fight against obesity and diabetes. Cyclic adenosine monophosphate (cAMP) is very important in the development of the beige phenotype and activation of its thermogenic program. To study effects of cyclic nucleotides on energy homeostatic mechanisms, mice were generated by targeted inactivation of cyclic nucleotide phosphodiesterase 3b (Pde3b) gene, which encodes PDE3B, an enzyme that catalyzes hydrolysis of cAMP and cGMP and is highly expressed in tissues that regulate energy homeostasis, including adipose tissue, liver, and pancreas. In epididymal white adipose tissue (eWAT) of PDE3B KO mice on a SvJ129 background, cAMP/protein kinase A (PKA) and AMP-activated protein kinase (AMPK) signaling pathways are activated, resulting in “browning” phenotype, with a smaller increases in body weight under high-fat diet, smaller fat deposits, increased β-oxidation of fatty acids (FAO) and oxygen consumption. Results reported here suggest that PDE3B and/or its downstream signaling partners might be important regulators of energy metabolism in adipose tissue, and potential therapeutic targets for treating obesity, diabetes and their associated metabolic disorders.
Collapse
Affiliation(s)
- Youn Wook Chung
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA.,Severance Integrative Research Institute for Cerebral and Cardiovascular Diseases (SIRIC), Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Faiyaz Ahmad
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Yan Tang
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Steven C Hockman
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Hyun Jung Kee
- Department of Surgery, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Karin Berger
- Lund University Diabetes Center, Department of Experimental Medical Sciences, Lund University, S-221 84 Lund, Sweden
| | - Emilia Guirguis
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Young Hun Choi
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Dan M Schimel
- NIH MRI Research Facility, NIH, Bethesda, Maryland, 20892, USA
| | - Angel M Aponte
- Proteomics Core Facility, NHLBI, NIH, Bethesda, Maryland, 20892, USA
| | - Sunhee Park
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Eva Degerman
- Lund University Diabetes Center, Department of Experimental Medical Sciences, Lund University, S-221 84 Lund, Sweden
| | - Vincent C Manganiello
- Cardiovascular and Pulmonary Branch (CPB), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| |
Collapse
|
56
|
Glöde A, Naumann J, Gnad T, Cannone V, Kilic A, Burnett JC, Pfeifer A. Divergent effects of a designer natriuretic peptide CD-NP in the regulation of adipose tissue and metabolism. Mol Metab 2017; 6:276-287. [PMID: 28271034 PMCID: PMC5323888 DOI: 10.1016/j.molmet.2016.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/20/2016] [Accepted: 12/29/2016] [Indexed: 01/06/2023] Open
Abstract
Objective Obesity is defined as an abnormal increase in white adipose tissue (WAT) and is a major risk factor for type 2 diabetes and cardiovascular disease. Brown adipose tissue (BAT) dissipates energy and correlates with leanness. Natriuretic peptides have been shown to be beneficial for brown adipocyte differentiation and browning of WAT. Methods Here, we investigated the effects of an optimized designer natriuretic peptide (CD-NP) on murine adipose tissues in vitro and in vivo. Results In murine brown and white adipocytes, CD-NP activated cGMP production, promoted adipogenesis, and increased thermogenic markers. Consequently, mice treated for 10 days with CD-NP exhibited increased “browning” of WAT. To study CD-NP effects on diet-induced obesity (DIO), we delivered CD-NP for 12 weeks. Although CD-NP reduced inflammation in WAT, CD-NP treated DIO mice exhibited a significant increase in body mass, worsened glucose tolerance, and hepatic steatosis. Long-term CD-NP treatment resulted in an increased expression of the NP scavenging receptor (NPR-C) and decreased lipolytic activity. Conclusions NP effects differed depending on the duration of treatment raising questions about the rational of natriuretic peptide treatment in obese patients. The optimized designer natriuretic peptide CD-NP promotes adipogenesis. Duration of treatment is decisive: short-term promotes browning whereas long-term treatment exacerbates obesity and diabetes. Long-term CD-NP treatment reduces WAT inflammation and increases adiponectin expression.
Collapse
Affiliation(s)
- Anja Glöde
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany; Bonn International Graduate School of Drug Sciences BIGSDrugS, University of Bonn, Bonn, Germany
| | - Jennifer Naumann
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Valentina Cannone
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, USA
| | - Ana Kilic
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, USA
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany; Bonn International Graduate School of Drug Sciences BIGSDrugS, University of Bonn, Bonn, Germany.
| |
Collapse
|
57
|
Effect of phosphodiesterase-5 inhibitors on glycemic control in person with type 2 diabetes mellitus: A systematic review and meta-analysis. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY 2016; 6:50-55. [PMID: 29067241 PMCID: PMC5644434 DOI: 10.1016/j.jcte.2016.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/16/2016] [Accepted: 11/16/2016] [Indexed: 12/30/2022]
Abstract
Chronic use of phosphodiesterase-5 inhibitors (PDE-5i) has been shown to improve insulin action on muscle glucose uptake by the prolongation of nitric oxide (NO)/cyclic guanosine monophosphate (cGMP)/protein kinase (PKG) signalling. Aims As the effects of PDE-5i on glycemic control in person with type 2 diabetes mellitus (T2DM) have not been systematically explored, we conducted a meta-analysis of available randomized controlled trials (RCTs). Methods A literature search was performed through electronic databases including MEDLINE (Pubmed), The Cochrane Library, SCOPUS, Web of Science, CINAHL, www.clinicaltrials.gov and www.clinicaltrialresults.org until April 2016 without language restriction. Studies were included if they met the following criteria: (i) RCTs of the chronic use of PDE-5i compared with placebo or no active treatment in T2DM patients (ii) reporting of HbA1c or glycated haemoglobin or fasting plasma glucose (FPG). Results Four studies involving a total of 198 patients fit into the inclusion criteria. All included studies used the same PDE-5i, sildenafil. Reports of HbA1c were analysed as only one study reported FPG. PDE-5i had no beneficial effect on HbA1c with weighted mean difference (WMD) of 0.17% (95% CI, −0.64 to 0.97). Conclusion This meta-analysis suggests that large and well-controlled studies are warranted to shed light on the effect of PDE-5i on glycemic control in people with type 2 diabetes mellitus.
Collapse
|
58
|
Aldiss P, Davies G, Woods R, Budge H, Sacks HS, Symonds ME. 'Browning' the cardiac and peri-vascular adipose tissues to modulate cardiovascular risk. Int J Cardiol 2016; 228:265-274. [PMID: 27865196 PMCID: PMC5236060 DOI: 10.1016/j.ijcard.2016.11.074] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/05/2016] [Indexed: 01/02/2023]
Abstract
Excess visceral adiposity, in particular that located adjacent to the heart and coronary arteries is associated with increased cardiovascular risk. In the pathophysiological state, dysfunctional adipose tissue secretes an array of factors modulating vascular function and driving atherogenesis. Conversely, brown and beige adipose tissues utilise glucose and lipids to generate heat and are associated with improved cardiometabolic health. The cardiac and thoracic perivascular adipose tissues are now understood to be composed of brown adipose tissue in the healthy state and undergo a brown-to-white transition i.e. during obesity which may be a driving factor of cardiovascular disease. In this review we discuss the risks of excess cardiac and vascular adiposity and potential mechanisms by which restoring the brown phenotype i.e. “re-browning” could potentially be achieved in clinically relevant populations. Epicardial, paracardial and thoracic perivascular adipose tissues resemble BAT at birth. Despite ‘whitening’ in early life these depots remain metabolically active and potentially thermogenic into adulthood. Obesity induces further ‘whitening’ and inflammation in these depots likely driving the atherogenesis. Maintaining or inducing the brown phenotype in these depots could prevent atherosclerotic disease.
Collapse
Affiliation(s)
- Peter Aldiss
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Graeme Davies
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Rachel Woods
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Helen Budge
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Harold S Sacks
- VA Greater Los Angeles Healthcare System, Endocrinology and Diabetes Division, and Department of Medicine David Geffen School of Medicine, Los Angeles, CA 90073, USA
| | - Michael E Symonds
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH.
| |
Collapse
|
59
|
Cameron RB, Beeson CC, Schnellmann RG. Development of Therapeutics That Induce Mitochondrial Biogenesis for the Treatment of Acute and Chronic Degenerative Diseases. J Med Chem 2016; 59:10411-10434. [PMID: 27560192 DOI: 10.1021/acs.jmedchem.6b00669] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondria have various roles in cellular metabolism and homeostasis. Because mitochondrial dysfunction is associated with many acute and chronic degenerative diseases, mitochondrial biogenesis (MB) is a therapeutic target for treating such diseases. Here, we review the role of mitochondrial dysfunction in acute and chronic degenerative diseases and the cellular signaling pathways by which MB is induced. We then review existing work describing the development and application of drugs that induce MB in vitro and in vivo. In particular, we discuss natural products and modulators of transcription factors, kinases, cyclic nucleotides, and G protein-coupled receptors.
Collapse
Affiliation(s)
- Robert B Cameron
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , 280 Calhoun Street, Charleston, South Carolina 29425, United States.,College of Pharmacy, University of Arizona , 1295 N. Martin Avenue, Tucson, Arizona 85721, United States
| | - Craig C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , 280 Calhoun Street, Charleston, South Carolina 29425, United States
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina , 280 Calhoun Street, Charleston, South Carolina 29425, United States.,College of Pharmacy, University of Arizona , 1295 N. Martin Avenue, Tucson, Arizona 85721, United States
| |
Collapse
|
60
|
Rixecker T, Mathar I, Medert R, Mannebach S, Pfeifer A, Lipp P, Tsvilovskyy V, Freichel M. TRPM4-mediated control of FcεRI-evoked Ca(2+) elevation comprises enhanced plasmalemmal trafficking of TRPM4 channels in connective tissue type mast cells. Sci Rep 2016; 6:32981. [PMID: 27624684 PMCID: PMC5021962 DOI: 10.1038/srep32981] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 08/17/2016] [Indexed: 11/09/2022] Open
Abstract
TRPM4 proteins form Ca2+-activated non selective cation (CAN) channels that affect transmembrane Ca2+-influx by determining the membrane potential. Tight control of the intracellular Ca2+ concentration is essential for mast cell responses. In this study, we analyzed the expression of TRPM4 in peritoneal mast cells (PCMC) as a model for connective tissue type mast cells with respect to FcεRI-evoked calcium changes and the subcellular localization of fluorescently labeled TRPM4 using two viral transduction systems before and following antigen stimulation. Our results show that TRPM4 is expressed in PCMCs, is an essential constituent of the endogenous CAN channels in PCMCs and regulates antigen-evoked increases in intracellular calcium that are significantly enhanced in TRPM4-deficient PCMCs. Compared to PCMCs analyzed before antigen stimulation, the cells depict a substantially increased localization of TRPM4 proteins towards the plasma membrane after FcεRI stimulation. Thus, TRPM4 functions as a limiting factor for antigen evoked calcium rise in connective tissue type mast cells and concurrent translocation of TRPM4 into the plasma membrane is part of this mechanism.
Collapse
Affiliation(s)
- Torben Rixecker
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Ilka Mathar
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Rebekka Medert
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Stefanie Mannebach
- Experimentelle und Klinische Pharmakologie und Toxikologie, Universität des Saarlandes, 66421 Homburg, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Peter Lipp
- Institut für Molekulare Zellbiologie Universität des Saarlandes, 66421 Homburg, Germany
| | - Volodymyr Tsvilovskyy
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| | - Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
61
|
Balkow A, Hoffmann LS, Klepac K, Glöde A, Gnad T, Zimmermann K, Pfeifer A. Direct lentivirus injection for fast and efficient gene transfer into brown and beige adipose tissue. J Biol Methods 2016; 3:e48. [PMID: 31453213 PMCID: PMC6706150 DOI: 10.14440/jbm.2016.123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/23/2016] [Accepted: 06/27/2016] [Indexed: 11/23/2022] Open
Abstract
Brown adipose tissue is a special type of fat contributing to energy expenditure in human newborns and adults. Moreover, subcutaneous white adipose tissue has a high capacity to adapt an energy-consuming, brown-like/beige phenotype. Here, we developed an easy to handle and fast to accomplish method to efficiently transfer genes into brown and beige fat pads in vivo. Lentiviral vectors are directly injected into the target fat pad of anesthetized mice through a small incision using a modified, small needle connected to a microsyringe, which is well suited for infiltration of adipose tissues. Expression of the target gene can be detected in brown/beige fat one week after injection. The method can be applied within minutes to efficiently deliver transgenes into subcutaneous adipose tissues. Thus, this protocol allows for studying genes of interest in a timely manner in murine brown/beige fat and could potentially lead to new gene therapies for obesity.
Collapse
Affiliation(s)
- Aileen Balkow
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Linda S Hoffmann
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Katarina Klepac
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany.,Research Training Group 1873, University of Bonn, 53127 Bonn, Germany
| | - Anja Glöde
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany.,BIGS DrugS International Graduate School, University of Bonn, 53127 Bonn, Germany
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Katrin Zimmermann
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany.,Research Training Group 1873, University of Bonn, 53127 Bonn, Germany.,BIGS DrugS International Graduate School, University of Bonn, 53127 Bonn, Germany.,PharmaCenter, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
62
|
Hankir MK, Kranz M, Gnad T, Weiner J, Wagner S, Deuther-Conrad W, Bronisch F, Steinhoff K, Luthardt J, Klöting N, Hesse S, Seibyl JP, Sabri O, Heiker JT, Blüher M, Pfeifer A, Brust P, Fenske WK. A novel thermoregulatory role for PDE10A in mouse and human adipocytes. EMBO Mol Med 2016; 8:796-812. [PMID: 27247380 PMCID: PMC4931292 DOI: 10.15252/emmm.201506085] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Phosphodiesterase type 10A (PDE10A) is highly enriched in striatum and is under evaluation as a drug target for several psychiatric/neurodegenerative diseases. Preclinical studies implicate PDE10A in the regulation of energy homeostasis, but the mechanisms remain unclear. By utilizing small-animal PET/MRI and the novel radioligand [(18)F]-AQ28A, we found marked levels of PDE10A in interscapular brown adipose tissue (BAT) of mice. Pharmacological inactivation of PDE10A with the highly selective inhibitor MP-10 recruited BAT and potentiated thermogenesis in vivo In diet-induced obese mice, chronic administration of MP-10 caused weight loss associated with increased energy expenditure, browning of white adipose tissue, and improved insulin sensitivity. Analysis of human PET data further revealed marked levels of PDE10A in the supraclavicular region where brown/beige adipocytes are clustered in adults. Finally, the inhibition of PDE10A with MP-10 stimulated thermogenic gene expression in human brown adipocytes and induced browning of human white adipocytes. Collectively, our findings highlight a novel thermoregulatory role for PDE10A in mouse and human adipocytes and promote PDE10A inhibitors as promising candidates for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany
| | - Mathias Kranz
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Neuroradiopharmaceuticals, Leipzig, Germany
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital University of Bonn, Bonn, Germany
| | - Juliane Weiner
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany
| | - Sally Wagner
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Neuroradiopharmaceuticals, Leipzig, Germany
| | - Winnie Deuther-Conrad
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Neuroradiopharmaceuticals, Leipzig, Germany
| | - Felix Bronisch
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany
| | - Karen Steinhoff
- Department of Nuclear Medicine, University Hospital University of Leipzig, Leipzig, Germany
| | - Julia Luthardt
- Department of Nuclear Medicine, University Hospital University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany
| | - Swen Hesse
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany Department of Nuclear Medicine, University Hospital University of Leipzig, Leipzig, Germany
| | | | - Osama Sabri
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany Department of Nuclear Medicine, University Hospital University of Leipzig, Leipzig, Germany
| | - John T Heiker
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital University of Bonn, Bonn, Germany
| | - Peter Brust
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf Neuroradiopharmaceuticals, Leipzig, Germany
| | - Wiebke K Fenske
- Integrated Research and Treatment Centre for Adiposity Diseases, University Hospital University of Leipzig, Leipzig, Germany
| |
Collapse
|
63
|
Abstract
The second messenger cyclic guanosine monophosphate (cGMP) is a key mediator in physiological processes such as vascular tone, and its essential involvement in pathways regulating metabolism has been recognized in recent years. Here, we focus on the fundamental role of cGMP in brown adipose tissue (BAT) differentiation and function. In contrast to white adipose tissue (WAT), which stores energy in the form of lipids, BAT consumes energy stored in lipids to generate heat. This so-called non-shivering thermogenesis takes place in BAT mitochondria, which express the specific uncoupling protein 1 (UCP1). The energy combusting properties of BAT render it a promising target in antiobesity strategies in which BAT could burn the surplus energy that has accumulated in obese and overweight individuals. cGMP is generated by guanylyl cyclases upon activation by nitric oxide or natriuretic peptides. It affects several downstream molecules including cGMP-receptor proteins such as cGMP-dependent protein kinase and is degraded by phosphodiesterases. The cGMP pathway contains several signaling molecules that can increase cGMP signaling, resulting in activation and recruitment of brown adipocytes, and hence can enhance the energy combusting features of BAT. In this review we highlight recent results showing the physiological significance of cGMP signaling in BAT, as well as pharmacological options targeting cGMP signaling that bear a high potential to become BAT-centered therapies for the treatment of obesity.
Collapse
|
64
|
Abstract
Cyclic guanosine 3′,5′-monophosphate (cGMP) serves as a second messenger molecule, which regulates pleiotropic cellular functions in health and disease. cGMP is generated by particulate or soluble guanylyl cyclases upon stimulation with natriuretic peptides or nitric oxide, respectively. Furthermore, the cGMP concentration is modulated by cGMP-degrading phosphodiesterases. Several targets of cGMP are utilized to effect its various cellular functions. These effector molecules comprise cGMP-dependent protein kinases, ion channels, and phosphodiesterases. During the last decade, it emerged that cGMP is a novel drug target for the treatment of pulmonary and cardiovascular disorders. In this respect, several drugs were developed, which are now in clinical phase studies for, e.g., pulmonary hypertension or cardiovascular diseases. These new drugs act NO-independently with/without heme on soluble guanylyl cyclases or induce subtypes of particular guanylyl cyclases and thereby lead to new therapeutic concepts and horizons. In this regard, the fifth cGMP meeting held in June 2011 in Halle, Germany, comprised the new therapeutic challenges with the novel functional and structural concepts of cGMP generating and effector molecules. This report summarizes the new data on molecular mechanisms, (patho)physiological relevance, and therapeutic potentials of the cGMP signaling system that were presented at this meeting.
Collapse
Affiliation(s)
- Jens Schlossmann
- Lehrstuhl für Pharmakologie und Toxikologie, Institut für Pharmazie, Universität Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Elisabeth Schinner
- Lehrstuhl für Pharmakologie und Toxikologie, Institut für Pharmazie, Universität Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| |
Collapse
|
65
|
Natriuretic peptide control of energy balance and glucose homeostasis. Biochimie 2016; 124:84-91. [DOI: 10.1016/j.biochi.2015.05.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/19/2015] [Indexed: 12/27/2022]
|
66
|
Exosomal microRNA miR-92a concentration in serum reflects human brown fat activity. Nat Commun 2016; 7:11420. [PMID: 27117818 PMCID: PMC4853423 DOI: 10.1038/ncomms11420] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/24/2016] [Indexed: 12/29/2022] Open
Abstract
Brown adipose tissue (BAT) dissipates energy and its activity correlates with leanness in human adults. 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography coupled with computer tomography (PET/CT) is still the standard for measuring BAT activity, but exposes subjects to ionizing radiation. To study BAT function in large human cohorts, novel diagnostic tools are needed. Here we show that brown adipocytes release exosomes and that BAT activation increases exosome release. Profiling miRNAs in exosomes released from brown adipocytes, and in exosomes isolated from mouse serum, we show that levels of miRNAs change after BAT activation in vitro and in vivo. One of these exosomal miRNAs, miR-92a, is also present in human serum exosomes. Importantly, serum concentrations of exosomal miR-92a inversely correlate with human BAT activity measured by 18F-FDG PET/CT in two unique and independent cohorts comprising 41 healthy individuals. Thus, exosomal miR-92a represents a potential serum biomarker for BAT activity in mice and humans. Exosomes are RNA-containing lipid vesicles with roles in inter-tissue crosstalk. Here the authors show that exosome release from brown adipocytes is increased upon thermogenic activation, both in vitro and in vivo, and demonstrate that serum levels of exosomal miR-92 reflect brown fat activity in humans.
Collapse
|
67
|
Maneschi E, Cellai I, Aversa A, Mello T, Filippi S, Comeglio P, Bani D, Guasti D, Sarchielli E, Salvatore G, Morelli A, Mazzanti B, Corcetto F, Corno C, Francomano D, Galli A, Vannelli GB, Lenzi A, Mannucci E, Maggi M, Vignozzi L. Tadalafil reduces visceral adipose tissue accumulation by promoting preadipocytes differentiation towards a metabolically healthy phenotype: Studies in rabbits. Mol Cell Endocrinol 2016; 424:50-70. [PMID: 26805634 DOI: 10.1016/j.mce.2016.01.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/18/2016] [Accepted: 01/18/2016] [Indexed: 12/15/2022]
Abstract
Development of metabolically healthy adipocytes within dysfunctional adipose tissue may represent an attractive way to counteract metabolic syndrome (MetS). In an experimental animal model of high fat diet (HFD)-induced MetS, in vivo, long- and short-term tadalafil treatments were able to reduce visceral adipose tissue (VAT) accumulation and hypertriglyceridemia, and to induce the expression in VAT of the brown fat-specific marker, uncoupling protein 1 (UCP1). VAT preadipocytes (PAD), isolated from the tadalafil-treated HFD rabbits, showed: i) a multilocular morphology; ii) an increased expression of brown fat-specific genes (such as UCP1 and CIDEA); iii) improved mitochondrial structure and dynamic and reduced superoxide production; iv) improved insulin sensitivity. Similar effects were obtained after in vitro tadalafil treatment in HFD rPAD. In conclusion, tadalafil counteracted HFD-associated VAT alterations, by restoring insulin-sensitivity and prompting preadipocytes differentiation towards a metabolically healthy phenotype.
Collapse
Affiliation(s)
- Elena Maneschi
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy
| | - Ilaria Cellai
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy
| | - Antonio Aversa
- Department of Experimental Medicine, Medical Pathophysiology, Food Science and Endocrinology Section, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Tommaso Mello
- Gastroenterology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Italy
| | - Sandra Filippi
- Interdepartmental Laboratory of Functional and Cellular Pharmacology of Reproduction, Department of Neuroscience, Drug Research and Child Care, Viale Pieraccini 6, 50139 University of Florence, Florence, Italy
| | - Paolo Comeglio
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy
| | - Daniele Bani
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, University of Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, University of Florence, Italy
| | - Erica Sarchielli
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, University of Florence, Italy
| | - Giulia Salvatore
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy
| | - Annamaria Morelli
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, University of Florence, Italy
| | - Benedetta Mazzanti
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, University of Florence, Italy
| | - Francesca Corcetto
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy
| | - Chiara Corno
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy
| | - Davide Francomano
- Department of Experimental Medicine, Medical Pathophysiology, Food Science and Endocrinology Section, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Andrea Galli
- Gastroenterology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Italy
| | - Gabriella Barbara Vannelli
- Department of Experimental and Clinical Medicine, Largo Brambilla 3, 50134, University of Florence, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Medical Pathophysiology, Food Science and Endocrinology Section, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Edoardo Mannucci
- Diabetes Section Geriatric Unit, Department of Critical Care, Careggi Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Mario Maggi
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy
| | - Linda Vignozzi
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, Viale Morgagni 50, 50134, University of Florence, Florence, Italy.
| |
Collapse
|
68
|
The Gq signalling pathway inhibits brown and beige adipose tissue. Nat Commun 2016; 7:10895. [PMID: 26955961 PMCID: PMC4786868 DOI: 10.1038/ncomms10895] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/01/2016] [Indexed: 12/31/2022] Open
Abstract
Brown adipose tissue (BAT) dissipates nutritional energy as heat via the uncoupling protein-1 (UCP1) and BAT activity correlates with leanness in human adults. Here we profile G protein-coupled receptors (GPCRs) in brown adipocytes to identify druggable regulators of BAT. Twenty-one per cent of the GPCRs link to the Gq family, and inhibition of Gq signalling enhances differentiation of human and murine brown adipocytes. In contrast, activation of Gq signalling abrogates brown adipogenesis. We further identify the endothelin/Ednra pathway as an autocrine activator of Gq signalling in brown adipocytes. Expression of a constitutively active Gq protein in mice reduces UCP1 expression in BAT, whole-body energy expenditure and the number of brown-like/beige cells in white adipose tissue (WAT). Furthermore, expression of Gq in human WAT inversely correlates with UCP1 expression. Thus, our data indicate that Gq signalling regulates brown/beige adipocytes and inhibition of Gq signalling may be a novel therapeutic approach to combat obesity.
Collapse
|
69
|
Musclin is an activity-stimulated myokine that enhances physical endurance. Proc Natl Acad Sci U S A 2015; 112:16042-7. [PMID: 26668395 DOI: 10.1073/pnas.1514250112] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Exercise remains the most effective way to promote physical and metabolic wellbeing, but molecular mechanisms underlying exercise tolerance and its plasticity are only partially understood. In this study we identify musclin-a peptide with high homology to natriuretic peptides (NP)-as an exercise-responsive myokine that acts to enhance exercise capacity in mice. We use human primary myoblast culture and in vivo murine models to establish that the activity-related production of musclin is driven by Ca(2+)-dependent activation of Akt1 and the release of musclin-encoding gene (Ostn) transcription from forkhead box O1 transcription factor inhibition. Disruption of Ostn and elimination of musclin secretion in mice results in reduced exercise tolerance that can be rescued by treatment with recombinant musclin. Reduced exercise capacity in mice with disrupted musclin signaling is associated with a trend toward lower levels of plasma atrial NP (ANP) and significantly smaller levels of cyclic guanosine monophosphate (cGMP) and peroxisome proliferator-activated receptor gamma coactivator 1-α in skeletal muscles after exposure to exercise. Furthermore, in agreement with the established musclin ability to interact with NP clearance receptors, but not with NP guanyl cyclase-coupled signaling receptors, we demonstrate that musclin enhances cGMP production in cultured myoblasts only when applied together with ANP. Elimination of the activity-related musclin-dependent boost of ANP/cGMP signaling results in significantly lower maximum aerobic capacity, mitochondrial protein content, respiratory complex protein expression, and succinate dehydrogenase activity in skeletal muscles. Together, these data indicate that musclin enhances physical endurance by promoting mitochondrial biogenesis.
Collapse
|
70
|
Sanyal A, Hoffmann LS, Etzrodt J, Pfeifer A. Effects of obesity on sGCβ1 mediated signaling in white adipose tissue. BMC Pharmacol Toxicol 2015. [PMCID: PMC4565486 DOI: 10.1186/2050-6511-16-s1-a83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
71
|
Roberts LD. Does inorganic nitrate say NO to obesity by browning white adipose tissue? Adipocyte 2015; 4:311-4. [PMID: 26451288 DOI: 10.1080/21623945.2015.1005525] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/02/2015] [Accepted: 01/05/2015] [Indexed: 12/28/2022] Open
Abstract
The dietary constituent inorganic nitrate, found in large concentrations in green leafy vegetables, has beneficial effects on cardiometabolic health. Contemporary studies employing nitrate have demonstrated that the anion has anti-obesity and anti-diabetic properties; however the nitrate-mediated mechanisms for improving metabolic health remain unclear. Recently, we employed a combined histological, metabolomics, and transcriptional and protein analysis approach to establish that nitrate promoted the "browning" of white adipose tissue via the xanthine oxidoreductase catalyzed reductive nitrate-nitrite-nitric oxide pathway. Interestingly, it was observed that nitrate-stimulated brown adipose-associated gene expression in white adipose tissue was augmented in hypoxia. These findings not only suggest that protection from metabolic disease offered by vegetable consumption may, in part, be mediated through the effects of nitrate on white adipose tissue, but also, since hypoxia is a serious co-morbidity affecting adipose tissue in obese individuals, that nitrate may be effective in promoting the browning of adipose tissue to improve metabolic fitness.
Collapse
|
72
|
Merlin J, Evans BA, Dehvari N, Sato M, Bengtsson T, Hutchinson DS. Could burning fat start with a brite spark? Pharmacological and nutritional ways to promote thermogenesis. Mol Nutr Food Res 2015. [DOI: 10.1002/mnfr.201500251] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jon Merlin
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
| | - Bronwyn A. Evans
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
| | - Nodi Dehvari
- Department of Molecular Biosciences; The Wenner-Gren Institute; Stockholm University; Stockholm Sweden
| | - Masaaki Sato
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
- Department of Pharmacology; Monash University; Clayton Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences; The Wenner-Gren Institute; Stockholm University; Stockholm Sweden
| | - Dana S. Hutchinson
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
- Department of Pharmacology; Monash University; Clayton Australia
| |
Collapse
|
73
|
Johlfs MG, Gorjala P, Urasaki Y, Le TT, Fiscus RR. Capillary Isoelectric Focusing Immunoassay for Fat Cell Differentiation Proteomics. PLoS One 2015; 10:e0132105. [PMID: 26132171 PMCID: PMC4489199 DOI: 10.1371/journal.pone.0132105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/10/2015] [Indexed: 01/09/2023] Open
Abstract
Profiling cellular proteome is critical to understanding signal integration during cell fate determination. In this study, the capability of capillary isoelectric focusing (cIEF) immunoassays to detect post-translational modifications (PTM) of protein isoforms is demonstrated. cIEF immunoassays exhibit protein detection sensitivity at up to 5 orders of magnitude higher than traditional methods. This detection ultra-sensitivity permits proteomic profiling of several nanograms of tissue samples. cIEF immunoassays are employed to simultaneously profile three protein kinases during fat cell differentiation: cGMP-dependent protein kinase type I (PKG-I) of the nitric oxide (NO) signaling pathway, protein kinase B (Akt) of the insulin signaling pathway, and extracellular signal-regulated kinase (ERK) of the mitogen-activated protein kinase (MAPK) signaling pathway. Interestingly, a switch in the expression level of PKG- isoforms is observed during fat cell differentiation. While both PKG-Iα and PKG-Iβ isoforms are present in preadipocytes, only PKG-Iβ isoform is expressed in adipocytes. On the other hand, the phosphorylation level increases for Akt while decreases for ERK1 and ERK2 following the maturation of preadipocytes into adipocytes. Taken together, cIEF immunoassay provides a highly sensitive means to study fat cell differentiation proteomics. cIEF immunoassay should be a powerful proteomics tool to study complex protein signal integration in biological systems.
Collapse
Affiliation(s)
- Mary G. Johlfs
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
| | - Priyatham Gorjala
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
| | - Yasuyo Urasaki
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
| | - Thuc T. Le
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
- * E-mail: (TTL); (RRF)
| | - Ronald R. Fiscus
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
- * E-mail: (TTL); (RRF)
| |
Collapse
|
74
|
A novel crosstalk between Alk7 and cGMP signaling differentially regulates brown adipocyte function. Mol Metab 2015; 4:576-83. [PMID: 26266090 PMCID: PMC4529496 DOI: 10.1016/j.molmet.2015.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 05/31/2015] [Accepted: 06/05/2015] [Indexed: 12/18/2022] Open
Abstract
Objective Obesity is an enormous burden for patients and health systems world-wide. Brown adipose tissue dissipates energy in response to cold and has been shown to be metabolically active in human adults. The type I transforming growth factor β (TGFβ) receptor Activin receptor-like kinase 7 (Alk7) is highly expressed in adipose tissues and is down-regulated in obese patients. Here, we studied the function of Alk7 in brown adipocytes. Methods Using pharmacological and genetic tools, Alk7 signaling pathway and its effects were studied in murine brown adipocytes. Brown adipocyte differentiation and activation was analyzed. Results Alk7 is highly upregulated during differentiation of brown adipocytes. Interestingly, Alk7 expression is increased by cGMP/protein kinase G (PKG) signaling, which enhances brown adipocyte differentiation. Activin AB effectively activates Alk7 and SMAD3 signaling. Activation of Alk7 in brown preadipocytes suppresses the master adipogenic transcription factor PPARγ and differentiation. Stimulation of Alk7 during late differentiation of brown adipocytes reduces lipid content and adipogenic marker expression but enhances UCP1 expression. Conclusions We found a so far unknown crosstalk between cGMP and Alk7 signaling pathways. Tight regulation of Alk7 is required for efficient differentiation of brown adipocytes. Alk7 has differential effects on adipogenic differentiation and the development of the thermogenic program in brown adipocytes. Alk7 expression in brown adipocytes is modulated by the cGMP/PKGI signaling pathway. Brown adipocyte differentiation requires tight regulation of Alk7 activation. Alk7 signaling via SMAD3 reduces PPARγ and attenuates adipogenic differentiation. Alk7 enhances UCP1 expression in mature brown adipocytes. We found a novel crosstalk between the Alk7 and cGMP signaling pathway.
Collapse
|
75
|
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: 84] [Impact Index Per Article: 8.4] [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
|
76
|
Maimaitiyiming H, Norman H, Zhou Q, Wang S. CD47 deficiency protects mice from diet-induced obesity and improves whole body glucose tolerance and insulin sensitivity. Sci Rep 2015; 5:8846. [PMID: 25747123 PMCID: PMC4352923 DOI: 10.1038/srep08846] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/05/2015] [Indexed: 02/03/2023] Open
Abstract
CD47 is a transmembrane protein with several functions including self-recognition, immune cell communication, and cell signaling. Although it has been extensively studied in cancer and ischemia, CD47 function in obesity has never been explored. In this study, we utilized CD47 deficient mice in a high-fat diet induced obesity model to study for the first time whether CD47 plays a role in the development of obesity and metabolic complications. Male CD47 deficient and wild type (WT) control mice were fed with either low fat (LF) or high fat (HF) diets for 16 weeks. Interestingly, we found that CD47 deficient mice were protected from HF diet-induced obesity displaying decreased weight gain and reduced adiposity. This led to decreased MCP1/CCR2 dependent macrophage infiltration into adipose tissue and reduced inflammation, resulting in improved glucose tolerance and insulin sensitivity. In addition, CD47 deficiency stimulated the expression of UCP1 and carnitine palmitoyltransferase 1b (CPT1b) levels in brown adipose tissue, leading to increased lipid utilization and heat production. This contributes to the increased energy utilization and reduced adiposity observed in these mice. Taken together, these data revealed a novel role for CD47 in the development of obesity and its related metabolic complications.
Collapse
Affiliation(s)
- Hasiyeti Maimaitiyiming
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536; Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Heather Norman
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536; Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Qi Zhou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536; Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536; Lexington Veterans Affairs Medical Center, Lexington, KY
| |
Collapse
|
77
|
Roberts LD, Ashmore T, Kotwica AO, Murfitt SA, Fernandez BO, Feelisch M, Murray AJ, Griffin JL. Inorganic nitrate promotes the browning of white adipose tissue through the nitrate-nitrite-nitric oxide pathway. Diabetes 2015; 64:471-484. [PMID: 25249574 PMCID: PMC4351918 DOI: 10.2337/db14-0496] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Inorganic nitrate was once considered an oxidation end product of nitric oxide metabolism with little biological activity. However, recent studies have demonstrated that dietary nitrate can modulate mitochondrial function in man and is effective in reversing features of the metabolic syndrome in mice. Using a combined histological, metabolomics, and transcriptional and protein analysis approach, we mechanistically defined that nitrate not only increases the expression of thermogenic genes in brown adipose tissue but also induces the expression of brown adipocyte-specific genes and proteins in white adipose tissue, substantially increasing oxygen consumption and fatty acid β-oxidation in adipocytes. Nitrate induces these phenotypic changes through a mechanism distinct from known physiological small molecule activators of browning, the recently identified nitrate-nitrite-nitric oxide pathway. The nitrate-induced browning effect was enhanced in hypoxia, a serious comorbidity affecting white adipose tissue in obese individuals, and corrected impaired brown adipocyte-specific gene expression in white adipose tissue in a murine model of obesity. Because resulting beige/brite cells exhibit antiobesity and antidiabetic effects, nitrate may be an effective means of inducing the browning response in adipose tissue to treat the metabolic syndrome.
Collapse
MESH Headings
- Adipocytes, Brown/physiology
- Adipocytes, White/drug effects
- Adipocytes, White/physiology
- Adipose Tissue, Brown
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Animals
- Cells, Cultured
- Cyclic GMP
- Cyclic GMP-Dependent Protein Kinases
- Dose-Response Relationship, Drug
- Male
- Mice
- Mice, Inbred C57BL
- Nitrates/metabolism
- Nitrates/pharmacology
- Nitric Oxide/metabolism
- Nitrites/metabolism
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- Lee D Roberts
- Medical Research Council – Human Nutrition Research, Elsie Widdowson Laboratory, 120 Fulbourn Road, Cambridge, CB2 9NL, UK
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Old Addenbrooke’s Site, Cambridge, CB2 1GA, UK
| | - Tom Ashmore
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Old Addenbrooke’s Site, Cambridge, CB2 1GA, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Aleksandra O Kotwica
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Steven A Murfitt
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Old Addenbrooke’s Site, Cambridge, CB2 1GA, UK
| | - Bernadette O Fernandez
- Faculty of Medicine, Clinical & Experimental Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Martin Feelisch
- Faculty of Medicine, Clinical & Experimental Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Julian L Griffin
- Medical Research Council – Human Nutrition Research, Elsie Widdowson Laboratory, 120 Fulbourn Road, Cambridge, CB2 9NL, UK
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, 80 Tennis Court Road, Old Addenbrooke’s Site, Cambridge, CB2 1GA, UK
| |
Collapse
|
78
|
Koksharova EO, Mayorov AY, Shestakova MV, Dedov II. Metabolic characteristics and therapeutic potential of brown and ?beige? adipose tissues. DIABETES MELLITUS 2014. [DOI: 10.14341/dm201445-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
According to the International Diabetes Federation, 10.9 million people have diabetes mellitus (DM) in Russia; however, only up to 4 million are registered. In addition, 11.9 million people have impaired glucose tolerance and impaired fasting glucose levels [1]. One of the significant risk factors for type 2 DM (T2DM) is obesity, which increases insulin resistance (IR). IR is the major pathogenetic link to T2DM. According to current concepts, there are three types of adipose tissue: white adipose tissue (WAT), brown adipose tissue (BAT) and ?beige?, of which the last two types have a thermogenic function. Some research results have revealed the main stages in the development of adipocytes; however, there is no general consensus regarding the development of ?beige? adipocytes. Furthermore, the biology of BAT and ?beige? adipose tissue is currently being intensively investigated, and some key transcription factors, signalling pathways and hormones that promote the development and activation of these tissues have been identified. The most discussed hormones are irisin and fibroblast growth factor 21, which have established positive effects on BAT and ?beige? adipose tissue with regard to carbohydrate, lipid and energy metabolism. The primary imaging techniques used to investigate BAT are PET-CT with 18F-fluorodeoxyglucose and magnetic resonance spectroscopy. With respect to the current obesity epidemic and associated diseases, including T2DM, there is a growing interest in investigating adipogenesis and the possibility of altering this process. BAT and ?beige? adipose tissue may be targets for developing drugs directed against obesity and T2DM.
Collapse
|
79
|
Mössenböck K, Vegiopoulos A, Rose AJ, Sijmonsma TP, Herzig S, Schafmeier T. Browning of white adipose tissue uncouples glucose uptake from insulin signaling. PLoS One 2014; 9:e110428. [PMID: 25313899 PMCID: PMC4197027 DOI: 10.1371/journal.pone.0110428] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/12/2014] [Indexed: 01/28/2023] Open
Abstract
Presence of thermogenically active adipose tissue in adult humans has been inversely associated with obesity and type 2 diabetes. While it had been shown that insulin is crucial for the development of classical brown fat, its role in development and function of inducible brown-in-white (brite) adipose tissue is less clear. Here we show that insulin deficiency impaired differentiation of brite adipocytes. However, adrenergic stimulation almost fully induced the thermogenic program under these settings. Although brite differentiation of adipocytes as well as browning of white adipose tissue entailed substantially elevated glucose uptake by adipose tissue, the capacity of insulin to stimulate glucose uptake surprisingly was not higher in the brite state. Notably, in line with the insulin-independent stimulation of glucose uptake, our data revealed that brite recruitment results in induction of solute carrier family 2 (GLUT-1) expression in adipocytes and inguinal WAT. These results for the first time demonstrate that insulin signaling is neither essential for brite recruitment, nor is it improved in cells or tissues upon browning.
Collapse
Affiliation(s)
- Karin Mössenböck
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, Heidelberg, Germany
| | - Alexandros Vegiopoulos
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, Heidelberg, Germany
- Junior research group Metabolism and Stem Cell Plasticity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adam J. Rose
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, Heidelberg, Germany
| | - Tjeerd P. Sijmonsma
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, Heidelberg, Germany
| | - Stephan Herzig
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, Heidelberg, Germany
- * E-mail:
| | - Tobias Schafmeier
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
80
|
Leiss V, Illison J, Domes K, Hofmann F, Lukowski R. Expression of cGMP-dependent protein kinase type I in mature white adipocytes. Biochem Biophys Res Commun 2014; 452:151-6. [DOI: 10.1016/j.bbrc.2014.08.071] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 08/15/2014] [Indexed: 01/01/2023]
|
81
|
MicroRNA-378 controls classical brown fat expansion to counteract obesity. Nat Commun 2014; 5:4725. [PMID: 25145289 PMCID: PMC4167820 DOI: 10.1038/ncomms5725] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/17/2014] [Indexed: 12/30/2022] Open
Abstract
Both classical brown adipocytes and brown-like beige adipocytes are considered as promising therapeutic targets for obesity; however, their development, relative importance, and functional coordination are not well understood. Here we show that a modest expression of miR-378/378* in adipose tissue specifically increases classical brown fat (BAT) mass, but not white fat (WAT) mass. Remarkably, BAT expansion, rather than miR-378 per se, suppresses formation of beige adipocytes in subcutaneous WAT. Despite this negative feedback, the expanded BAT depot is sufficient to prevent both genetic and high fat diet-induced obesity. At the molecular level, we find that miR-378 targets phosphodiesterase Pde1b in BAT, but not in WAT. Indeed, miR-378 and Pde1b inversely regulate brown adipogenesis in vitro in the absence of phosphodiesterase inhibitor IBMX. Our work identifies miR-378 as a key regulatory component underlying classical BAT-specific expansion and obesity resistance, and adds novel insights into the physiological cross-talk between BAT and WAT.
Collapse
|
82
|
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: 118] [Impact Index Per Article: 10.7] [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
|
83
|
Kasza I, Suh Y, Wollny D, Clark RJ, Roopra A, Colman RJ, MacDougald OA, Shedd TA, Nelson DW, Yen MI, Yen CLE, Alexander CM. Syndecan-1 is required to maintain intradermal fat and prevent cold stress. PLoS Genet 2014; 10:e1004514. [PMID: 25101993 PMCID: PMC4125098 DOI: 10.1371/journal.pgen.1004514] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 06/05/2014] [Indexed: 02/07/2023] Open
Abstract
Homeostatic temperature regulation is fundamental to mammalian physiology and is controlled by acute and chronic responses of local, endocrine and nervous regulators. Here, we report that loss of the heparan sulfate proteoglycan, syndecan-1, causes a profoundly depleted intradermal fat layer, which provides crucial thermogenic insulation for mammals. Mice without syndecan-1 enter torpor upon fasting and show multiple indicators of cold stress, including activation of the stress checkpoint p38α in brown adipose tissue, liver and lung. The metabolic phenotype in mutant mice, including reduced liver glycogen, is rescued by housing at thermoneutrality, suggesting that reduced insulation in cool temperatures underlies the observed phenotypes. We find that syndecan-1, which functions as a facultative lipoprotein uptake receptor, is required for adipocyte differentiation in vitro. Intradermal fat shows highly dynamic differentiation, continuously expanding and involuting in response to hair cycle and ambient temperature. This physiology probably confers a unique role for Sdc1 in this adipocyte sub-type. The PPARγ agonist rosiglitazone rescues Sdc1−/− intradermal adipose tissue, placing PPARγ downstream of Sdc1 in triggering adipocyte differentiation. Our study indicates that disruption of intradermal adipose tissue development results in cold stress and complex metabolic pathology. All mammals strive to maintain a fixed body temperature, and do so using a remarkable array of different strategies, which vary depending upon the degree of cold challenge. Physiologists many decades ago observed that a fat layer right underneath the epidermis (and above the dermal muscle layer) thickens in response to colder ambient temperatures. This “intradermal fat” provided insulation within days of climate changes. We have found that syndecan-1, which functions as a facultative lipoprotein uptake receptor, is required for intradermal fat expansion in response to cold exposure. This is a highly specific phenotype not shared by other adipocytes. When intradermal fat is absent, mice do not adapt normally to cold stress, and show altered systemic physiologies, including increased brown adipose tissue thermogenesis and hyper-activation of a stress checkpoint (p38α), designed to protect the body against mutagenic and oxidative stressors. The phenotypes associated with loss of Sdc1 function are reversed when mice are housed in warm temperatures, where defense of body temperature is not required. This study is the first to show that intradermal fat can be genetically regulated, with systemic effects on physiology.
Collapse
Affiliation(s)
- Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Yewseok Suh
- Department of Dermatology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Damian Wollny
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rod J. Clark
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ricki J. Colman
- Wisconsin National Primate Research Center, Madison, Wisconsin, United States of America
| | - Ormond A. MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Timothy A. Shedd
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David W. Nelson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mei-I Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Chi-Liang Eric Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Caroline M. Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
84
|
Hoffmann LS, Chen HH. cGMP: transition from bench to bedside: a report of the 6th International Conference on cGMP Generators, Effectors and Therapeutic Implications. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:707-18. [PMID: 24927824 DOI: 10.1007/s00210-014-0999-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 05/30/2014] [Indexed: 02/06/2023]
Abstract
Essential physiological homeostatic processes such as vascular tone, fluid balance, cardiorenal function, and sensory processes are regulated by the second messenger cyclic guanosine 3', 5'-monophosphate (cGMP). Dysregulation of cGMP-dependent pathways plays an important role in cardiovascular diseases such as hypertension, pulmonary hypertension, heart failure, or erectile dysfunction. Thus, the cGMP pathway consisting of the cGMP-generating guanylyl cyclases, protein kinases, and phosphodiesterases (PDE) has evolved to an important drug target and is the focus of a wide variety of research fields ranging from unraveling mechanisms on the molecular level to understanding the regulation of physiological and pathophysiological processes by cGMP. Based on the results from basic and preclinical research, therapeutic drugs have been developed which modulate the cGMP pathway and are investigated in clinical trials. Riociguat, a nitric oxide (NO)-independent soluble guanylyl cyclase stimulator; recombinant B-type natriuretic peptide (BNP); or recombinant atrial natriuretic peptide (ANP) and PDE5 inhibitors are cGMP-modulating drugs that are already available for the treatment of pulmonary hypertension, acute heart failure, and erectile dysfunction, respectively. The latest results from basic to clinical research on cGMP were presented on the 6th International Conference on cGMP in Erfurt, Germany, and are summarized in this article.
Collapse
Affiliation(s)
- Linda S Hoffmann
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, Bonn, Germany,
| | | |
Collapse
|
85
|
Li C, Sun D, Zhang S, Wang S, Wu X, Zhang Q, Liu L, Li Y, Qiao L. Genome wide association study identifies 20 novel promising genes associated with milk fatty acid traits in Chinese Holstein. PLoS One 2014; 9:e96186. [PMID: 24858810 PMCID: PMC4032272 DOI: 10.1371/journal.pone.0096186] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 04/03/2014] [Indexed: 12/11/2022] Open
Abstract
Detecting genes associated with milk fat composition could provide valuable insights into the complex genetic networks of genes underling variation in fatty acids synthesis and point towards opportunities for changing milk fat composition via selective breeding. In this study, we conducted a genome-wide association study (GWAS) for 22 milk fatty acids in 784 Chinese Holstein cows with the PLINK software. Genotypes were obtained with the Illumina BovineSNP50 Bead chip and a total of 40,604 informative, high-quality single nucleotide polymorphisms (SNPs) were used. Totally, 83 genome-wide significant SNPs and 314 suggestive significant SNPs associated with 18 milk fatty acid traits were detected. Chromosome regions that affect milk fatty acid traits were mainly observed on BTA1, 2, 5, 6, 7, 9, 13, 14, 18, 19, 20, 21, 23, 26 and 27. Of these, 146 SNPs were associated with more than one milk fatty acid trait; most of studied fatty acid traits were significant associated with multiple SNPs, especially C18:0 (105 SNPs), C18 index (93 SNPs), and C14 index (84 SNPs); Several SNPs are close to or within the DGAT1, SCD1 and FASN genes which are well-known to affect milk composition traits of dairy cattle. Combined with the previously reported QTL regions and the biological functions of the genes, 20 novel promising candidates for C10:0, C12:0, C14:0, C14:1, C14 index, C18:0, C18:1n9c, C18 index, SFA, UFA and SFA/UFA were found, which composed of HTR1B, CPM, PRKG1, MINPP1, LIPJ, LIPK, EHHADH, MOGAT1, ECHS1, STAT1, SORBS1, NFKB2, AGPAT3, CHUK, OSBPL8, PRLR, IGF1R, ACSL3, GHR and OXCT1. Our findings provide a groundwork for unraveling the key genes and causal mutations affecting milk fatty acid traits in dairy cattle.
Collapse
Affiliation(s)
- Cong Li
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dongxiao Sun
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- * E-mail:
| | - Shengli Zhang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Sheng Wang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiaoping Wu
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qin Zhang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lin Liu
- Beijing Dairy Cattle Center, Beijing, China
| | - Yanhua Li
- Beijing Dairy Cattle Center, Beijing, China
| | - Lv Qiao
- Beijing Dairy Cattle Center, Beijing, China
| |
Collapse
|
86
|
Lee SH, Huang H, Choi K, Lee DH, Shi J, Liu T, Chun KH, Seo JA, Lima IS, Zabolotny JM, Wei L, Kim YB. ROCK1 isoform-specific deletion reveals a role for diet-induced insulin resistance. Am J Physiol Endocrinol Metab 2014; 306:E332-43. [PMID: 24326423 PMCID: PMC3920011 DOI: 10.1152/ajpendo.00619.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Rho kinase (ROCK) isoforms regulate insulin signaling and glucose metabolism negatively or positively in cultured cell lines and skeletal muscle. However, the in vivo function of the ROCK1 isoform in adipose tissue has not been addressed. To determine the specific role of the adipose ROCK1 isoform in the development of insulin resistance and obesity, mice lacking ROCK1 in adipose tissue globally or selectively were studied. Here, we show that insulin's ability to activate IRS-1/PI3K/Akt signaling was greatly enhanced in adipose tissue of ROCK1(-/-) mice compared with wild-type mice. These effects resulted from the inhibitory effect of ROCK1 on insulin receptor action, as evidenced by the fact that IR tyrosine phosphorylation was abolished in ROCK1(-/-) MEF cells when ROCK1 was reexpressed. Consistently, adipose-specific disruption of ROCK1 increased IR tyrosine phosphorylation in adipose tissue and modestly improved sensitivity to insulin in obese mice induced by high-fat feeding. This effect is independent of any changes in adiposity, number or size of adipocytes, and metabolic parameters, including glucose, insulin, leptin, and triglyceride levels, demonstrating a minimal effect of adipose ROCK1 on whole body metabolism. Enzymatic activity of ROCK1 in adipose tissue remained ∼50%, which likely originated from the fraction of stromal vascular cells, suggesting involvement of these cells for adipose metabolic regulation. Moreover, ROCK isoform activities were increased in adipose tissue of diet-induced or genetically obese mice. These data suggest that adipose ROCK1 isoform plays an inhibtory role for the regulation of insulin sensitivity in diet-induced obesity in vivo.
Collapse
Affiliation(s)
- Seung-Hwan Lee
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Colombo G, Colombo MDHP, Schiavon LDL, d'Acampora AJ. Phosphodiesterase 5 as target for adipose tissue disorders. Nitric Oxide 2013; 35:186-192. [PMID: 24177060 DOI: 10.1016/j.niox.2013.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 09/26/2013] [Accepted: 10/21/2013] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Adipose tissue as an endocrine organ is responsible for the release of multiple cytokines, which have the most diverse metabolic functions. Therefore, it is extremely important to preserve its physiological health in order to avoid local and systemic disorders. Experiments available in literature show the importance of the nitric oxide (NO)/guanosine 3'5' cyclic monophosphate (cGMP)/protein kinase G (PKG) pathway in adipocyte biology. Phosphodiesterase 5 (PDE5) is an enzyme responsible for cGMP inactivation, and the use of its inhibitors can be an alternative in the search of a more balanced adipose tissue. OBJECTIVE This review aims to describe the PDE5 role and the possibility of using PDE5 inhibitors in adipocyte physiology derangements and their consequences. DESIGN AND METHODS Studies published in the last 10years that related PDE5 and its inhibitors to adipose tissue were raised in major databases. RESULTS PDE5 is present in adipocyte, and PDE5 inhibitors can promote adipogenesis, interfere with adipokines secretion, decrease inflammatory markers expression, and increase the thermogenic potential of white adipose tissue. CONCLUSIONS PDE5 plays an important role in adipocyte physiology and the use of its inhibitors may prove a useful tool to combat adipose tissue disorders and its highest expression, metabolic syndrome.
Collapse
|
88
|
miR-155 regulates differentiation of brown and beige adipocytes via a bistable circuit. Nat Commun 2013; 4:1769. [PMID: 23612310 PMCID: PMC3644088 DOI: 10.1038/ncomms2742] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 03/15/2013] [Indexed: 12/11/2022] Open
Abstract
Brown adipocytes are a primary site of energy expenditure and reside not only in classical brown adipose tissue but can also be found in white adipose tissue. Here we show that microRNA 155 is enriched in brown adipose tissue and is highly expressed in proliferating brown preadipocytes but declines after induction of differentiation. Interestingly, microRNA 155 and its target, the adipogenic transcription factor CCAAT/enhancer-binding protein β, form a bistable feedback loop integrating hormonal signals that regulate proliferation or differentiation. Inhibition of microRNA 155 enhances brown adipocyte differentiation and induces a brown adipocyte-like phenotype (‘browning’) in white adipocytes. Consequently, microRNA 155-deficient mice exhibit increased brown adipose tissue function and ‘browning’ of white fat tissue. In contrast, transgenic overexpression of microRNA 155 in mice causes a reduction of brown adipose tissue mass and impairment of brown adipose tissue function. These data demonstrate that the bistable loop involving microRNA 155 and CCAAT/enhancer-binding protein β regulates brown lineage commitment, thereby, controlling the development of brown and beige fat cells. Brown fat can dissipate energy as heat and has an important role in energy homoeostasis of rodents and possibly humans. Chen et al. show that microRNA 155 regulates the differentiation of brown adipocytes as well as the 'browning' of white fat cells in mice.
Collapse
|
89
|
Kajimura S, Saito M. A new era in brown adipose tissue biology: molecular control of brown fat development and energy homeostasis. Annu Rev Physiol 2013; 76:225-49. [PMID: 24188710 DOI: 10.1146/annurev-physiol-021113-170252] [Citation(s) in RCA: 331] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Brown adipose tissue (BAT) is specialized to dissipate chemical energy in the form of heat as a defense against cold and excessive feeding. Interest in the field of BAT biology has exploded in the past few years because of the therapeutic potential of BAT to counteract obesity and obesity-related diseases, including insulin resistance. Much progress has been made, particularly in the areas of BAT physiology in adult humans, developmental lineages of brown adipose cell fate, and hormonal control of BAT thermogenesis. As we enter into a new era of brown fat biology, the next challenge will be to develop strategies for activating BAT thermogenesis in adult humans to increase whole-body energy expenditure. This article reviews the recent major advances in this field and discusses emerging questions.
Collapse
Affiliation(s)
- Shingo Kajimura
- Diabetes Center, Department of Cell and Tissue Biology, University of California, San Francisco, California 94143-0669;
| | | |
Collapse
|
90
|
Kim GW, Lin JE, Blomain ES, Waldman SA. Antiobesity pharmacotherapy: new drugs and emerging targets. Clin Pharmacol Ther 2013; 95:53-66. [PMID: 24105257 DOI: 10.1038/clpt.2013.204] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 09/18/2013] [Indexed: 02/07/2023]
Abstract
Obesity is a growing pandemic, and related health and economic costs are staggering. Pharmacotherapy, partnered with lifestyle modifications, forms the core of current strategies to reduce the burden of this disease and its sequelae. However, therapies targeting weight loss have a significant history of safety risks, including cardiovascular and psychiatric events. Here, evolving strategies for developing antiobesity therapies, including targets, mechanisms, and developmental status, are highlighted. Progress in this field is underscored by Belviq (lorcaserin) and Qsymia (phentermine/topiramate), the first agents in more than 10 years to achieve regulatory approval for chronic weight management in obese patients. On the horizon, novel insights into metabolism and energy homeostasis reveal guanosine 3',5'-cyclic monophosphate (cGMP) signaling circuits as emerging targets for antiobesity pharmacotherapy. These innovations in molecular discovery may elegantly align with practical off-the-shelf approaches, leveraging existing approved drugs that modulate cGMP levels for the management of obesity.
Collapse
Affiliation(s)
- G W Kim
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - J E Lin
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - E S Blomain
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - S A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
91
|
|
92
|
Sanyal A, Lochmatter D, Preußner A, Pfeifer A. Influence of PKG on insulin signalling and GSK3 phosphorylation in SH-SY5Y cells. BMC Pharmacol Toxicol 2013. [PMCID: PMC3765466 DOI: 10.1186/2050-6511-14-s1-p60] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
93
|
Etzrodt J, Hoffmann LS, Friebe A, Pfeifer A. The soluble guanylyl cyclase stimulator BAY 41-2272 increases differentiation and function of brown adipocytes. BMC Pharmacol Toxicol 2013. [PMCID: PMC3765490 DOI: 10.1186/2050-6511-14-s1-p20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
94
|
Maimaitiyiming H, Li Y, Cui W, Tong X, Norman H, Qi X, Wang S. Increasing cGMP-dependent protein kinase I activity attenuates cisplatin-induced kidney injury through protection of mitochondria function. Am J Physiol Renal Physiol 2013; 305:F881-90. [PMID: 23825069 DOI: 10.1152/ajprenal.00192.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cisplatin is widely used to treat malignancies. However, its major limitation is the development of dose-dependent nephrotoxicity. The precise mechanisms of cisplatin-induced kidney damage remain unclear, and the renoprotective agents during cisplatin treatment are still lacking. Here, we demonstrated that the expression and activity of cGMP-dependent protein kinase-I (PKG-I) were reduced in cisplatin-treated renal tubular cells in vitro as well as in the kidney tissues from cisplatin-treated mice in vivo. Increasing PKG activity by both pharmacological and genetic approaches attenuated cisplatin-induced kidney cell apoptosis in vitro. This was accompanied by decreased Bax/Bcl2 ratio, caspase 3 activity, and cytochrome c release. Cisplatin-induced mitochondria membrane potential loss in the tubular cells was also prevented by increased PKG activity. All of these data suggest a protective effect of PKG on mitochondria function in renal tubular cells. Importantly, increasing PKG activity pharmacologically or genetically diminished cisplatin-induced tubular damage and preserved renal function during cisplatin treatment in vivo. Mitochondria structural and functional damage in the kidney from cisplatin-treated mice was inhibited by increased PKG activity. In addition, increasing PKG activity enhanced ciaplatin-induced cell death in several cancer cell lines. Taken together, these results suggest that increasing PKG activity may be a novel option for renoprotection during cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Hasiyeti Maimaitiyiming
- Graduate Center for Nutritional Sciences, Univ. of Kentucky, Wethington Bldg. Rm. 583, 900 S. Limestone St., Lexington, KY 40536.
| | | | | | | | | | | | | |
Collapse
|
95
|
Pfeifer A, Kilić A, Hoffmann LS. Regulation of metabolism by cGMP. Pharmacol Ther 2013; 140:81-91. [PMID: 23756133 DOI: 10.1016/j.pharmthera.2013.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 01/16/2023]
Abstract
The second messenger cyclic guanosine monophosphate (cGMP) mediates the physiological effects of nitric oxide and natriuretic peptides in a broad spectrum of tissues and cells. So far, the major focus of research on cGMP lay on the cardiovascular system. Recent evidence suggests that cGMP also plays a major role in the regulation of cellular and whole-body metabolism. Here, we focus on the role of cGMP in adipose tissue. In addition, other organs important for the regulation of metabolism and their regulation by cGMP are discussed. Targeting the cGMP signaling pathway could be an exciting approach for the regulation of energy expenditure and the treatment of obesity.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, Germany.
| | | | | |
Collapse
|
96
|
Rohm M, Sommerfeld A, Strzoda D, Jones A, Sijmonsma TP, Rudofsky G, Wolfrum C, Sticht C, Gretz N, Zeyda M, Leitner L, Nawroth PP, Stulnig TM, Berriel Diaz M, Vegiopoulos A, Herzig S. Transcriptional cofactor TBLR1 controls lipid mobilization in white adipose tissue. Cell Metab 2013; 17:575-85. [PMID: 23499424 DOI: 10.1016/j.cmet.2013.02.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 12/14/2012] [Accepted: 02/05/2013] [Indexed: 11/30/2022]
Abstract
Lipid mobilization (lipolysis) in white adipose tissue (WAT) critically controls lipid turnover and adiposity in humans. While the acute regulation of lipolysis has been studied in detail, the transcriptional determinants of WAT lipolytic activity remain still largely unexplored. Here we show that the genetic inactivation of transcriptional cofactor transducin beta-like-related 1(TBLR1) blunts the lipolytic response of white adipocytes through the impairment of cAMP-dependent signal transduction. Indeed, mice lacking TBLR1 in adipocytes are defective in fasting-induced lipid mobilization and, when placed on a high-fat-diet, show aggravated adiposity, glucose intolerance, and insulin resistance. TBLR1 levels are found to increase under lipolytic conditions in WAT of both human patients and mice, correlating with serum free fatty acids (FFAs). As a critical regulator of WAT cAMP signaling and lipid mobilization, proper activity of TBLR1 in adipocytes might thus represent a critical molecular checkpoint for the prevention of metabolic dysfunction in subjects with obesity-related disorders.
Collapse
Affiliation(s)
- Maria Rohm
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH) and University Hospital, Heidelberg University, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Abstract
During the last decades, obesity research has focused on food intake regulation, whereas energy expenditure has been mainly measured based on whole-body oxygen consumption. With the renaissance of brown adipose tissue (BAT) thermogenesis as a potential drug target in humans, more thought is put into alternative heat-producing mechanisms. Also, the interaction of peripheral and central components to regulate thermogenesis requires further studies. Certainly, several of the novel molecular genetic tools available now, compared with 40 years ago, will be helpful to gain new insights in BAT-controlled energy homeostasis and promises new approaches to pharmacologically control body weight.
Collapse
Affiliation(s)
- Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany.
| | | |
Collapse
|
98
|
Mitschke MM, Hoffmann LS, Gnad T, Scholz D, Kruithoff K, Mayer P, Haas B, Sassmann A, Pfeifer A, Kilic A. Increased cGMP promotes healthy expansion and browning of white adipose tissue. FASEB J 2013; 27:1621-30. [PMID: 23303211 DOI: 10.1096/fj.12-221580] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
With more than half a billion individuals affected worldwide, obesity has reached pandemic proportions. Development of "brown-like" or "brite" adipocytes within white adipose tissue (WAT) has potential antiobesity and insulin-sensitizing effects. We investigated the role of cyclic GMP (cGMP) signaling, focusing on cGMP-dependent protein kinase I (PKGI) in WAT. PKGI is expressed in murine WAT, primary adipocytes, and 3T3-L1. Treatment of adipocytes with cGMP resulted in increased adipogenesis, with a 54% increase in expression of peroxisome proliferator-activated receptor-γ. Lentiviral overexpression of PKGI further increased adipogenesis, whereas loss of PKGI significantly reduced adipogenic differentiation. In addition to adipogenic effects, PKGI had an antihypertrophic and anti-inflammatory effect via RhoA phosphorylation and reduction of proinflammatory adipokine expression. Moreover, PKGI induced a 4.3-fold increase in abundance of UCP-1 and the development of a brown-like thermogenic program in primary adipocytes. Notably, treatment of C57BL/6 mice with phosphodiesterase inhibitor sildenafil (12 mg/kg/d) for 7 d caused 4.6-fold increase in uncoupling protein-1 expression and promoted establishment of a brown fat cell-like phenotype ("browning") of WAT in vivo. Taken together, PKGI is a key regulator of cell size, adipokine secretion and browning of white fat depots and thus could be a valuable target in developing novel treatments for obesity.
Collapse
|
99
|
Abstract
cGMP-dependent protein kinases (cGK) are serine/threonine kinases that are widely distributed in eukaryotes. Two genes-prkg1 and prkg2-code for cGKs, namely, cGKI and cGKII. In mammals, two isozymes, cGKIα and cGKIβ, are generated from the prkg1 gene. The cGKI isozymes are prominent in all types of smooth muscle, platelets, and specific neuronal areas such as cerebellar Purkinje cells, hippocampal neurons, and the lateral amygdala. The cGKII prevails in the secretory epithelium of the small intestine, the juxtaglomerular cells, the adrenal cortex, the chondrocytes, and in the nucleus suprachiasmaticus. Both cGKs are major downstream effectors of many, but not all, signalling events of the NO/cGMP and the ANP/cGMP pathways. cGKI relaxes smooth muscle tone and prevents platelet aggregation, whereas cGKII inhibits renin secretion, chloride/water secretion in the small intestine, the resetting of the clock during early night, and endochondral bone growth. This chapter focuses on the involvement of cGKs in cardiovascular and non-cardiovascular processes including cell growth and metabolism.
Collapse
Affiliation(s)
- Franz Hofmann
- FOR 923, Institut für Pharmakologie und Toxikologie, der Technischen Universität München, Munich, Germany
| | | |
Collapse
|
100
|
Jennissen K, Haas B, Mitschke MM, Siegel F, Pfeifer A. Analysis of cGMP signaling in adipocytes. Methods Mol Biol 2013; 1020:175-92. [PMID: 23709033 DOI: 10.1007/978-1-62703-459-3_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity has reached pandemic dimensions with more than half a billion adults affected worldwide. Detailed knowledge of adipose biology is required for the development of urgently needed novel therapies directed against obesity. Two types of adipose tissue can be distinguished in humans and mice: white adipose tissue (WAT), which primarily stores energy in the form of lipids and has endocrine functions. In contrast, brown adipose tissue (BAT) dissipates energy in the form of heat (thermogenesis). Recent studies in humans demonstrated that BAT not only plays a role for non-shivering thermogenesis in newborns but is also metabolically active in adults. Here, we describe protocols for the generation of cellular models for the analysis of adipogenesis as well as function of brown and white fat. These models are based on the in vitro differentiation of mesenchymal stem cells (MSCs) isolated from adipose tissues. Using specific differentiation protocols, the role of cGMP signaling in both brown as well as white adipocytes can be studied.
Collapse
Affiliation(s)
- Katja Jennissen
- Institute of Pharmacology and Toxicology, Universität Bonn, Bonn, Germany
| | | | | | | | | |
Collapse
|