1
|
Salicylate Sodium Suppresses Monocyte Chemoattractant Protein-1 Production by Directly Inhibiting Phosphodiesterase 3B in TNF-α-Stimulated Adipocytes. Int J Mol Sci 2022; 24:ijms24010320. [PMID: 36613764 PMCID: PMC9820166 DOI: 10.3390/ijms24010320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
As a worldwide health issue, obesity is associated with the infiltration of monocytes/macrophages into the adipose tissue causing unresolved inflammation. Monocyte chemoattractant protein-1 (MCP-1) exerts a crucial effect on obesity-related monocytes/macrophages infiltration. Clinically, aspirin and salsalate are beneficial for the treatment of metabolic diseases in which adipose tissue inflammation plays an essential role. Herein, we investigated the effect and precise mechanism of their active metabolite salicylate on TNF-α-elevated MCP-1 in adipocytes. The results indicated that salicylate sodium (SAS) could lower the level of MCP-1 in TNF-α-stimulated adipocytes, which resulted from a previously unrecognized target phosphodiesterase (PDE), 3B (PDE3B), rather than its known targets IKKβ and AMPK. The SAS directly bound to the PDE3B to inactivate it, thus elevating the intracellular cAMP level and activating PKA. Subsequently, the expression of MKP-1 was increased, which led to the decrease in p-EKR and p-p38. Both PDE3B silencing and the pharmacological inhibition of cAMP/PKA compromised the suppressive effect of SAS on MCP-1. In addition to PDE3B, the PDE3A and PDE4B activity was also inhibited by SAS. Our findings identify a previously unrecognized pathway through which SAS is capable of attenuating the inflammation of adipocytes.
Collapse
|
2
|
Medak KD, McKie GL, Shamshoum H, Seguin I, Wright DC. The glucose lowering effects of CL 316,243 dissipate with repeated use and are rescued bycilostamide. Physiol Rep 2022; 10:e15187. [PMID: 35179321 PMCID: PMC8855634 DOI: 10.14814/phy2.15187] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
Repeated activation of the beta 3 adrenergic receptor (β3AR) with the agonist CL 316,243 (CL) results in remodeling of white adipose tissue (WAT) characterized by increased mitochondrial enzymes and expression of uncoupling protein 1 (UCP1). β3AR activation also has profound acute metabolic effects including rapidly decreasing blood glucose, secondary to fatty acid-induced increases in insulin, and increasing energy expenditure. The acute (single dose) effects of β3AR activation have largely been examined in treatment naive animals and under room temperature housing conditions. The current study examined if repeated CL treatment would lead to an attenuation of acute metabolic effects of CL treatment under thermoneutral housing conditions and if this could be rescued with cilostamide, a phosphodiesterase inhibitor. We provide evidence demonstrating that the acute effects of CL to increase serum fatty acids and insulin and reduce blood glucose, but not increases in energy expenditure, are attenuated in mice following repeated treatment with CL. This occurs in parallel with reductions in indices of protein kinase A signaling in WAT including the phosphorylation of hormone sensitive lipase. The findings of attenuated serum fatty acid, insulin, and blood glucose responses were confirmed in both high-fat fed and UCP1-/- mice repeatedly treated with CL. Desensitization to CL in mice was rescued by cilostamide. Herein, we provide evidence that the glucose lowering, but not thermogenesis inducing, effects of CL are attenuated with repeated treatment and can be rescued by cilostamide. The findings of this study point toward novel adjunct treatment approaches that could be used to maximize therapeutic, glucose lowering effects of β3AR agonists.
Collapse
Affiliation(s)
- Kyle D. Medak
- Department of Human Health and Nutritional SciencesUniversity of GuelphGuelphOntarioCanada
| | - Greg L. McKie
- Department of Human Health and Nutritional SciencesUniversity of GuelphGuelphOntarioCanada
| | - Hesham Shamshoum
- Department of Human Health and Nutritional SciencesUniversity of GuelphGuelphOntarioCanada
| | - Ian Seguin
- Department of Human Health and Nutritional SciencesUniversity of GuelphGuelphOntarioCanada
| | - David C. Wright
- Department of Human Health and Nutritional SciencesUniversity of GuelphGuelphOntarioCanada
| |
Collapse
|
3
|
Campolo F, Pofi R, Venneri MA, Isidori AM. Priming metabolism with the type 5 phosphodiesterase: the role of cGMP-hydrolyzing enzymes. Curr Opin Pharmacol 2021; 60:298-305. [PMID: 34507030 DOI: 10.1016/j.coph.2021.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/24/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022]
Abstract
The cyclic guanosine monophosphate (cGMP) signaling system is one of the most prominent regulators of many physiopathological processes in humans and rodents. It has been strongly established as an accomplished cellular signal involved in the regulation of energy homeostasis and cell metabolism, and pharmacological enhancement of cGMP has shown beneficial effects in metabolic disorders models. cGMP intracellular levels are finely regulated by phosphodiesterases (PDEs). The main enzyme responsible for the degradation of cGMP is PDE5. Preclinical and clinical studies have shown that PDE5 inhibitors (PDE5i) have beneficial effects on improving insulin resistance and glucose metabolism representing a promising therapeutic strategy for the treatment of metabolic disorders. This review aims to describe the molecular basis underlying the use of PDE5i to prompt cell metabolism and summarize current clinical trials assessing the effects of PDE5i on glucose metabolism.
Collapse
Affiliation(s)
- Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Italy.
| |
Collapse
|
4
|
Ceddia RP, Collins S. A compendium of G-protein-coupled receptors and cyclic nucleotide regulation of adipose tissue metabolism and energy expenditure. Clin Sci (Lond) 2020; 134:473-512. [PMID: 32149342 PMCID: PMC9137350 DOI: 10.1042/cs20190579] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022]
Abstract
With the ever-increasing burden of obesity and Type 2 diabetes, it is generally acknowledged that there remains a need for developing new therapeutics. One potential mechanism to combat obesity is to raise energy expenditure via increasing the amount of uncoupled respiration from the mitochondria-rich brown and beige adipocytes. With the recent appreciation of thermogenic adipocytes in humans, much effort is being made to elucidate the signaling pathways that regulate the browning of adipose tissue. In this review, we focus on the ligand-receptor signaling pathways that influence the cyclic nucleotides, cAMP and cGMP, in adipocytes. We chose to focus on G-protein-coupled receptor (GPCR), guanylyl cyclase and phosphodiesterase regulation of adipocytes because they are the targets of a large proportion of all currently available therapeutics. Furthermore, there is a large overlap in their signaling pathways, as signaling events that raise cAMP or cGMP generally increase adipocyte lipolysis and cause changes that are commonly referred to as browning: increasing mitochondrial biogenesis, uncoupling protein 1 (UCP1) expression and respiration.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| |
Collapse
|
5
|
Yi X, Wu P, Liu J, Gong Y, Xu X, Li W. Identification of the potential key genes for adipogenesis from human mesenchymal stem cells by RNA-Seq. J Cell Physiol 2019; 234:20217-20227. [PMID: 30989650 DOI: 10.1002/jcp.28621] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/22/2019] [Indexed: 01/23/2023]
Abstract
Adipogenesis, a physiological process initiated with the committed preadipocytes expressing adipocyte-specific genes and terminated in mature, differentiated and functional adipocytes, mainly involved with energy homeostasis. Abnormal distribution-changes and dysfunctions in adipogenesis may lead to complex physiopathological disorders. However, it remains unclear for the key players working for the whole complex differentiating process of adipogenesis. Here, it investigated transcriptional profiling of adipogenesis from human mesenchymal stem cells (hMSCs) by RNA-Seq transcriptome technique. Oil Red O staining assays were performed to assess adipogenic potential. Quantitative real-time PCR (qRT-PCR) and lentivirus transfection assays by small interference RNA (siRNA) were conducted to confirm the function of the candidate genes. A total of 1,078 differentially expressed genes shared at 7, 14, 21, and 28 days during adipogenesis from hMSCs, and 706 genes were significantly differentially expressed. It identified 20 potential key genes responsible for adipogenesis with four genes downregulating. The candidate gene, coagulation factor II thrombin receptor (F2R), encoding coagulation factor II thrombin receptor involving with a 7-transmembrane receptor involved in the regulation of thrombotic response, also known as proteinase-activated receptor-1, contributed to adipogenesis, especially at Day 14, by Oil Red O staining, qRT-PCR, and western blot after siRNA. A unique discovery shed new light to understand the key players of the whole processes of adipogenesis from hMSCs. The gene F2R might be used as an adipogenic marker to provide a potential target for understanding the metabolic syndromes like obesity, type-2 diabetes, steatosis, atherosclerosis, and osteoporosis.
Collapse
Affiliation(s)
- Xia Yi
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, China
| | - Ping Wu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, China
| | - Jianyun Liu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, China
| | - Ying Gong
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, China
| | - Xiaoyuan Xu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, China
| | - Weidong Li
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, China
| |
Collapse
|
6
|
Varghese M, Griffin C, McKernan K, Eter L, Lanzetta N, Agarwal D, Abrishami S, Singer K. Sex Differences in Inflammatory Responses to Adipose Tissue Lipolysis in Diet-Induced Obesity. Endocrinology 2019; 160:293-312. [PMID: 30544158 PMCID: PMC6330175 DOI: 10.1210/en.2018-00797] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/09/2018] [Indexed: 02/08/2023]
Abstract
Males are known to have profound adipose tissue macrophage (ATM) accumulation in gonadal white adipose tissue (GWAT) during obesity, whereas females are protected from such an inflammatory response even with increased adiposity. The inflammatory tone in males is linked to insulin resistance and might be the underlying cause for sex differences in metabolic disease. Factors regulating the meta-inflammatory response remain unclear but enhanced lipid storage in females may explain the reduced inflammatory response to high-fat diets. In this study, we evaluated lean and obese females with stimulated lipolysis to understand whether a stress release of free fatty acids (FFAs) could induce female ATMs. We demonstrate that in both lean and obese females, GWAT CD11c- resident ATMs accumulate with β-3 adrenergic receptor-stimulated lipolysis. Lipolysis elevated serum FFA, triglyceride, and IL-6 levels in females that corresponded to significant phosphorylated hormone-sensitive lipase and adipose triglyceride lipase protein expression in obese female GWAT compared with males. Increased lipolytic response in obese females was associated with crown-like structures and induced Il6, Mcp1, Arg1, and Mgl1 expression in obese female GWAT, suggesting an environment of lipid clearance and adipose remodeling. With this finding we next investigated whether lipid storage and lipolytic mediators differed by sex. Diacylglycerol, ceramides, phospholipids, and certain fatty acid species associated with inflammation were elevated in male GWAT compared with obese female GWAT. Overall, our data demonstrate a role for GWAT lipid storage and lipolytic metabolites to induce inflammation in males and induce remodeling in females that might explain sex differences in overall metabolic health.
Collapse
Affiliation(s)
- Mita Varghese
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
| | - Cameron Griffin
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kaitlin McKernan
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
| | - Leila Eter
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
| | - Nicholas Lanzetta
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
| | - Devyani Agarwal
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
| | - Simin Abrishami
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kanakadurga Singer
- Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan
- Correspondence: Kanakadurga Singer, MD, Department of Pediatrics and Communicable Diseases, Division of Pediatric Endocrinology, D1205 MPB, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
7
|
Emdin CA, Khera AV, Chaffin M, Klarin D, Natarajan P, Aragam K, Haas M, Bick A, Zekavat SM, Nomura A, Ardissino D, Wilson JG, Schunkert H, McPherson R, Watkins H, Elosua R, Bown MJ, Samani NJ, Baber U, Erdmann J, Gupta N, Danesh J, Chasman D, Ridker P, Denny J, Bastarache L, Lichtman JH, D’Onofrio G, Mattera J, Spertus JA, Sheu WHH, Taylor KD, Psaty BM, Rich SS, Post W, Rotter JI, Chen YDI, Krumholz H, Saleheen D, Gabriel S, Kathiresan S. Analysis of predicted loss-of-function variants in UK Biobank identifies variants protective for disease. Nat Commun 2018; 9:1613. [PMID: 29691411 PMCID: PMC5915445 DOI: 10.1038/s41467-018-03911-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/21/2018] [Indexed: 02/02/2023] Open
Abstract
Less than 3% of protein-coding genetic variants are predicted to result in loss of protein function through the introduction of a stop codon, frameshift, or the disruption of an essential splice site; however, such predicted loss-of-function (pLOF) variants provide insight into effector transcript and direction of biological effect. In >400,000 UK Biobank participants, we conduct association analyses of 3759 pLOF variants with six metabolic traits, six cardiometabolic diseases, and twelve additional diseases. We identified 18 new low-frequency or rare (allele frequency < 5%) pLOF variant-phenotype associations. pLOF variants in the gene GPR151 protect against obesity and type 2 diabetes, in the gene IL33 against asthma and allergic disease, and in the gene IFIH1 against hypothyroidism. In the gene PDE3B, pLOF variants associate with elevated height, improved body fat distribution and protection from coronary artery disease. Our findings prioritize genes for which pharmacologic mimics of pLOF variants may lower risk for disease.
Collapse
Affiliation(s)
- Connor A. Emdin
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Amit V. Khera
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Mark Chaffin
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Derek Klarin
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA ,000000041936754Xgrid.38142.3cDepartment of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Pradeep Natarajan
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Krishna Aragam
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Mary Haas
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Alexander Bick
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Seyedeh M. Zekavat
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA ,0000000419368710grid.47100.32Department of Computational Biology & Bioinformatics, Yale Medical School, Yale University, New Haven, MA 06510 USA
| | - Akihiro Nomura
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Diego Ardissino
- grid.411482.aDivision of Cardiology, Azienda Ospedaliero–Universitaria di Parma, Parma, 43121 Italy ,Associazione per lo Studio Della Trombosi in Cardiologia, Pavia, 27100 Italy
| | - James G. Wilson
- 0000 0004 1937 0407grid.410721.1Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Heribert Schunkert
- 0000 0004 5937 5237grid.452396.fDeutsches Herzzentrum München, Technische Universität München, Deutsches Zentrum für Herz-Kreislauf-Forschung, München, 80333 Germany
| | - Ruth McPherson
- 0000 0001 2182 2255grid.28046.38University of Ottawa Heart Institute, Ottawa, ON K1Y4W7 Canada
| | - Hugh Watkins
- 0000 0004 1936 8948grid.4991.5Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, OX1 2JD UK ,0000 0004 1936 8948grid.4991.5Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX1 2JD UK
| | - Roberto Elosua
- 0000 0004 1767 8811grid.411142.3Cardiovascular Epidemiology and Genetics, Hospital del Mar Research Institute, Barcelona, 08003 Spain ,CIBER Enfermedades Cardiovasculares (CIBERCV), Barcelona, 28029 Spain ,Facultat de Medicina, Universitat de Vic-Central de Cataluña, Barcelona, VIC 08500 Spain
| | - Matthew J. Bown
- 0000 0004 1936 8411grid.9918.9Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Biomedical Research Centre, Leicester, LE1 7RH UK
| | - Nilesh J. Samani
- 0000 0004 1936 8411grid.9918.9Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Biomedical Research Centre, Leicester, LE1 7RH UK
| | - Usman Baber
- 0000 0001 0670 2351grid.59734.3cThe Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, 10029 NY USA
| | - Jeanette Erdmann
- 0000 0001 0057 2672grid.4562.5Institute for Integrative and Experimental Genomics, University of Lübeck, Lübeck, 23562 Germany
| | - Namrata Gupta
- grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - John Danesh
- 0000000121885934grid.5335.0Department of Public Health and Primary Care, Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB2 0SR UK ,0000 0004 0606 5382grid.10306.34Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA UK ,0000000121885934grid.5335.0National Institute of Health Research Blood and Transplant; Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, CB2 1TN UK
| | - Daniel Chasman
- 0000 0004 0378 8294grid.62560.37Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Boston, 02115 USA
| | - Paul Ridker
- 0000 0004 0378 8294grid.62560.37Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Boston, 02115 USA
| | - Joshua Denny
- 0000 0001 2264 7217grid.152326.1Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37235 USA
| | - Lisa Bastarache
- 0000 0001 2264 7217grid.152326.1Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37235 USA
| | - Judith H. Lichtman
- 0000000419368710grid.47100.32Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT 06510 USA
| | - Gail D’Onofrio
- 0000000419368710grid.47100.32Department of Emergency Medicine, Yale University, New Haven, CT 06520 USA
| | - Jennifer Mattera
- grid.417307.6Center for Outcomes Research and Evaluation, Yale–New Haven Hospital, New Haven, CT 06510 USA
| | - John A. Spertus
- 0000 0001 2179 926Xgrid.266756.6Department of Biomedical & Saint Luke’s Mid America Heart Institute and the Health Informatics, Division of Endocrinology and Metabolism, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Wayne H.-H. Sheu
- 0000 0004 0573 0731grid.410764.0Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, 40705 Taiwan
| | - Kent D. Taylor
- 0000 0001 0157 6501grid.239844.0The Institute for Translational Genomics and Population Sciences, LABioMed and Department of Pediatrics at Harbor-UCLA Medical Center, Torrance, CA 90095 USA
| | - Bruce M. Psaty
- 0000000122986657grid.34477.33Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, 98195 WA USA ,0000 0004 0615 7519grid.488833.cCardiovascular Health Research Unit, Kaiser Permanente Washington Health Research Institute, 98101 Seattle, WA USA
| | - Stephen S. Rich
- 0000 0000 9136 933Xgrid.27755.32Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA 22908 USA
| | - Wendy Post
- 0000 0001 2171 9311grid.21107.35Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Jerome I. Rotter
- 0000 0001 0157 6501grid.239844.0The Institute for Translational Genomics and Population Sciences, LABioMed and Department of Pediatrics at Harbor-UCLA Medical Center, Torrance, CA 90095 USA
| | - Yii-Der Ida Chen
- 0000 0001 0157 6501grid.239844.0The Institute for Translational Genomics and Population Sciences, LABioMed and Department of Pediatrics at Harbor-UCLA Medical Center, Torrance, CA 90095 USA
| | - Harlan Krumholz
- grid.417307.6Center for Outcomes Research and Evaluation, Yale–New Haven Hospital, New Haven, CT 06510 USA
| | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, 74800 Pakistan ,0000 0004 1936 8972grid.25879.31Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Stacey Gabriel
- grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Sekar Kathiresan
- 000000041936754Xgrid.38142.3cCenter for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,000000041936754Xgrid.38142.3cDepartment of Medicine, Massachusetts General Hospital, Cardiology Division, Harvard Medical School, Boston, MA 02114 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| |
Collapse
|
8
|
Dünnes S, Voussen B, Aue A, Groneberg K, Nikolaev V, Groneberg D, Friebe A. Phosphodiesterase 3A expression and activity in the murine vasculature is influenced by NO-sensitive guanylyl cyclase. Pflugers Arch 2018; 470:693-702. [PMID: 29294149 DOI: 10.1007/s00424-017-2106-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/15/2017] [Accepted: 12/21/2017] [Indexed: 11/27/2022]
Abstract
Phosphodiesterase 3 (PDE3) exists in two isoforms (PDE3A and PDE3B) and is known to act as cGMP-inhibited cAMP-degrading PDE. Therefore, PDE3 may likely be involved in the interaction between the two second messenger pathways. NO-sensitive guanylyl cyclase (NO-GC) is the most important cytosolic generator of cGMP. Here, we investigated the effect of NO-GC deletion on PDE3A-mediated signaling in animals lacking NO-GC either globally (GCKO) or specifically in smooth muscle cells (SMC-GCKO). PDE3A expression is detected in murine aortic smooth muscle, platelets, and heart tissue. Expression and activity of PDE3A in aortae from GCKO and SMC-GCKO mice was reduced by approx. 50% compared to that in control animals. PDE3A downregulation can be linked to the reduction in NO-GC and is not an effect of the increased blood pressure levels resulting from NO-GC deletion. Despite the different PDE3A expression levels, smooth muscle relaxation induced by forskolin to stimulate cAMP signaling was similar in all genotypes. Basal and forskolin-stimulated cAMP levels in aortic tissue were not different between KO and control strains. However, the potency of milrinone, a selective inhibitor of PDE3A, to induce relaxation was higher in aortae from GCKO and SMC-GCKO than that in aorta from control animals. These data were corroborated by the effect of milrinone in vivo, which led to an increase in systolic blood pressure in both KO strains but not in control mice. We conclude that NO-GC modulates PDE3A expression and activity in SMC in vivo conceivably to preserve functional cAMP signaling.
Collapse
Affiliation(s)
- Sarah Dünnes
- Physiologisches Institut I, Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Barbara Voussen
- Physiologisches Institut I, Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Annemarie Aue
- Physiologisches Institut I, Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Kaja Groneberg
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Viacheslav Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dieter Groneberg
- Physiologisches Institut I, Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany
| | - Andreas Friebe
- Physiologisches Institut I, Universität Würzburg, Röntgenring 9, 97070, Würzburg, Germany.
| |
Collapse
|
9
|
Hinney A, Kesselmeier M, Jall S, Volckmar AL, Föcker M, Antel J, Heid IM, Winkler TW, Grant SFA, Guo Y, Bergen AW, Kaye W, Berrettini W, Hakonarson H, Herpertz-Dahlmann B, de Zwaan M, Herzog W, Ehrlich S, Zipfel S, Egberts KM, Adan R, Brandys M, van Elburg A, Boraska Perica V, Franklin CS, Tschöp MH, Zeggini E, Bulik CM, Collier D, Scherag A, Müller TD, Hebebrand J. Evidence for three genetic loci involved in both anorexia nervosa risk and variation of body mass index. Mol Psychiatry 2017; 22:192-201. [PMID: 27184124 PMCID: PMC5114162 DOI: 10.1038/mp.2016.71] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 02/22/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023]
Abstract
The maintenance of normal body weight is disrupted in patients with anorexia nervosa (AN) for prolonged periods of time. Prior to the onset of AN, premorbid body mass index (BMI) spans the entire range from underweight to obese. After recovery, patients have reduced rates of overweight and obesity. As such, loci involved in body weight regulation may also be relevant for AN and vice versa. Our primary analysis comprised a cross-trait analysis of the 1000 single-nucleotide polymorphisms (SNPs) with the lowest P-values in a genome-wide association meta-analysis (GWAMA) of AN (GCAN) for evidence of association in the largest published GWAMA for BMI (GIANT). Subsequently we performed sex-stratified analyses for these 1000 SNPs. Functional ex vivo studies on four genes ensued. Lastly, a look-up of GWAMA-derived BMI-related loci was performed in the AN GWAMA. We detected significant associations (P-values <5 × 10-5, Bonferroni-corrected P<0.05) for nine SNP alleles at three independent loci. Interestingly, all AN susceptibility alleles were consistently associated with increased BMI. None of the genes (chr. 10: CTBP2, chr. 19: CCNE1, chr. 2: CARF and NBEAL1; the latter is a region with high linkage disequilibrium) nearest to these SNPs has previously been associated with AN or obesity. Sex-stratified analyses revealed that the strongest BMI signal originated predominantly from females (chr. 10 rs1561589; Poverall: 2.47 × 10-06/Pfemales: 3.45 × 10-07/Pmales: 0.043). Functional ex vivo studies in mice revealed reduced hypothalamic expression of Ctbp2 and Nbeal1 after fasting. Hypothalamic expression of Ctbp2 was increased in diet-induced obese (DIO) mice as compared with age-matched lean controls. We observed no evidence for associations for the look-up of BMI-related loci in the AN GWAMA. A cross-trait analysis of AN and BMI loci revealed variants at three chromosomal loci with potential joint impact. The chromosome 10 locus is particularly promising given that the association with obesity was primarily driven by females. In addition, the detected altered hypothalamic expression patterns of Ctbp2 and Nbeal1 as a result of fasting and DIO implicate these genes in weight regulation.
Collapse
Affiliation(s)
- A Hinney
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - M Kesselmeier
- Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - S Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - A-L Volckmar
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - M Föcker
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - J Antel
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - I M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - T W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - S F A Grant
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
- Divisions of Genetics and Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Y Guo
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - W Kaye
- Department of Psychiatry, University of California, San Diego, San Diego, CA, USA
| | - W Berrettini
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - H Hakonarson
- The Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - B Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of the RWTH Aachen, Aachen, Germany
| | - M de Zwaan
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - W Herzog
- Department of Internal Medicine II, General Internal and Psychosomatic Medicine, University of Heidelberg, Heidelberg, Germany
| | - S Ehrlich
- Translational Developmental Neuroscience Section, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU-Dresden, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - S Zipfel
- Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital, Tübingen, Germany
| | - K M Egberts
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - R Adan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- Altrecht Eating Disorders Rintveld, Zeist, The Netherlands
| | - M Brandys
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
- Altrecht Eating Disorders Rintveld, Zeist, The Netherlands
| | - A van Elburg
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - V Boraska Perica
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
- University of Split School of Medicine, Split, Croatia
| | - C S Franklin
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - E Zeggini
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - C M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - D Collier
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
- Eli Lilly and Company Ltd, Surrey, UK
| | - A Scherag
- Clinical Epidemiology, Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center & German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - J Hebebrand
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
10
|
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
|
11
|
Ponnusamy S, Tran QT, Harvey I, Smallwood HS, Thiyagarajan T, Banerjee S, Johnson DL, Dalton JT, Sullivan RD, Miller DD, Bridges D, Narayanan R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue. FASEB J 2016; 31:266-281. [PMID: 27733447 DOI: 10.1096/fj.201600787rr] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/22/2016] [Indexed: 01/03/2023]
Abstract
Most satiety-inducing obesity therapeutics, despite modest efficacy, have safety concerns that underscore the need for effective peripherally acting drugs. An attractive therapeutic approach for obesity is to optimize/maximize energy expenditure by increasing energy-utilizing thermogenic brown adipose tissue. We used in vivo and in vitro models to determine the role of estrogen receptor β (ER-β) and its ligands on adipose biology. RNA sequencing and metabolomics were used to determine the mechanism of action of ER-β and its ligands. Estrogen receptor β (ER-β) and its selective ligand reprogrammed preadipocytes and precursor stem cells into brown adipose tissue and increased mitochondrial respiration. An ER-β-selective ligand increased markers of tricarboxylic acid-dependent and -independent energy biogenesis and oxygen consumption in mice without a concomitant increase in physical activity or food consumption, all culminating in significantly reduced weight gain and adiposity. The antiobesity effects of ER-β ligand were not observed in ER-β-knockout mice. Serum metabolite profiles of adult lean and juvenile mice were comparable, while that of adult obese mice was distinct, indicating a possible impact of obesity on age-dependent metabolism. This phenotype was partially reversed by ER-β-selective ligand. These data highlight a new role for ER-β in adipose biology and its potential to be a safer alternative peripheral therapeutic target for obesity.-Ponnusamy, S., Tran, Q. T., Harvey, I., Smallwood, H. S., Thiyagarajan, T., Banerjee, S., Johnson, D. L., Dalton, J. T., Sullivan, R. D., Miller, D. D., Bridges, D., Narayanan, R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Quynh T Tran
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Innocence Harvey
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thirumagal Thiyagarajan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Daniel L Johnson
- Molecular Informatics Core, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James T Dalton
- Preclinical Research and Development, GTx, Incorporated, Memphis, Tennessee, USA
| | - Ryan D Sullivan
- Department of Comparative Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; and
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dave Bridges
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; .,West Cancer Center, Memphis, Tennessee, USA
| |
Collapse
|
12
|
Phosphodiesterase 3B (PDE3B) regulates NLRP3 inflammasome in adipose tissue. Sci Rep 2016; 6:28056. [PMID: 27321128 PMCID: PMC4913246 DOI: 10.1038/srep28056] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 05/31/2016] [Indexed: 11/25/2022] Open
Abstract
Activation of inflammation in white adipose tissue (WAT), includes infiltration/expansion of WAT macrophages, contributes pathogenesis of obesity, insulin resistance, and metabolic syndrome. The inflammasome comprises an intracellular sensor (NLR), caspase-1 and the adaptor ASC. Inflammasome activation leads to maturation of caspase-1 and processing of IL1β, contributing to many metabolic disorders and directing adipocytes to a more insulin-resistant phenotype. Ablation of PDE3B in WAT prevents inflammasome activation by reducing expression of NLRP3, caspase-1, ASC, AIM2, TNFα, IL1β and proinflammatory genes. Following IP injection of lipopolysaccharide (LPS), serum levels of IL1β and TNFα were reduced in PDE3B−/−mice compared to WT. Activation of signaling cascades, which mediate inflammasome responses, were modulated in PDE3B−/−mice WAT, including smad, NFAT, NFkB, and MAP kinases. Moreover, expression of chemokine CCL2, MCP-1 and its receptor CCR2, which play an important role in macrophage chemotaxis, were reduced in WAT of PDE3B−/−mice. In addition, atherosclerotic plaque formation was significantly reduced in the aorta of apoE−/−/PDE3B−/−and LDL-R−/−/PDE3B−/−mice compared to apoE−/−and LDL-R−/−mice, respectively. Obesity-induced changes in serum-cholesterol were blocked in PDE3B−/−mice. Collectively, these data establish a role for PDE3B in modulating inflammatory response, which may contribute to a reduced inflammatory state in adipose tissue.
Collapse
|
13
|
Ravnskjaer K, Madiraju A, Montminy M. Role of the cAMP Pathway in Glucose and Lipid Metabolism. Handb Exp Pharmacol 2016; 233:29-49. [PMID: 26721678 DOI: 10.1007/164_2015_32] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
3'-5'-Cyclic adenosine monophosphate (cyclic AMP or cAMP) was first described in 1957 as an intracellular second messenger mediating the effects of glucagon and epinephrine on hepatic glycogenolysis (Berthet et al., J Biol Chem 224(1):463-475, 1957). Since this initial characterization, cAMP has been firmly established as a versatile molecular signal involved in both central and peripheral regulation of energy homeostasis and nutrient partitioning. Many of these effects appear to be mediated at the transcriptional level, in part through the activation of the transcription factor CREB and its coactivators. Here we review current understanding of the mechanisms by which the cAMP signaling pathway triggers metabolic programs in insulin-responsive tissues.
Collapse
|
14
|
Ye H, Wang X, Sussman CR, Hopp K, Irazabal MV, Bakeberg JL, LaRiviere WB, Manganiello VC, Vorhees CV, Zhao H, Harris PC, van Deursen J, Ward CJ, Torres VE. Modulation of Polycystic Kidney Disease Severity by Phosphodiesterase 1 and 3 Subfamilies. J Am Soc Nephrol 2016; 27:1312-20. [PMID: 26374610 PMCID: PMC4849815 DOI: 10.1681/asn.2015010057] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 08/04/2015] [Indexed: 11/03/2022] Open
Abstract
Aberrant intracellular calcium levels and increased cAMP signaling contribute to the development of polycystic kidney disease (PKD). cAMP can be hydrolyzed by various phosphodiesterases (PDEs). To examine the role of cAMP hydrolysis and the most relevant PDEs in the pathogenesis of PKD, we examined cyst development in Pde1- or Pde3-knockout mice on the Pkd2(-/WS25) background (WS25 is an unstable Pkd2 allele). These PDEs were selected because of their importance in cross-talk between calcium and cyclic nucleotide signaling (PDE1), control of cell proliferation and cystic fibrosis transmembrane conductance regulator (CFTR) -driven fluid secretion (PDE3), and response to vasopressin V2 receptor activation (both). In Pkd2(-/WS25) mice, knockout of Pde1a, Pde1c, or Pde3a but not of Pde1b or Pde3b aggravated the development of PKD and was associated with higher levels of protein kinase A-phosphorylated (Ser133) cAMP-responsive binding protein (P-CREB), activating transcription factor-1, and CREB-induced CRE modulator proteins in kidney nuclear preparations. Immunostaining also revealed higher expression of P-CREB in Pkd2(-/) (WS25);Pde1a(-/-), Pkd2(-) (/WS25);Pde1c(-/-), and Pkd2(-/) (WS25);Pde3a(-/-) kidneys. The cystogenic effect of desmopressin administration was markedly enhanced in Pkd2(-/WS25);Pde3a(-/-) mice, despite PDE3 accounting for only a small fraction of renal cAMP PDE activity. These observations show that calcium- and calmodulin-dependent PDEs (PDE1A and PDE1C) and PDE3A modulate the development of PKD, possibly through the regulation of compartmentalized cAMP pools that control cell proliferation and CFTR-driven fluid secretion. Treatments capable of increasing the expression or activity of these PDEs may, therefore, retard the development of PKD.
Collapse
Affiliation(s)
- Hong Ye
- Division of Nephrology and Hypertension and
| | | | | | | | | | - Jason L Bakeberg
- Division of Nephrology and Hypertension, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | | | - Vincent C Manganiello
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland
| | - Charles V Vorhees
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Research Foundation and University of Cincinnati, Cincinnati, Ohio; and
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | | | - Jan van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Christopher J Ward
- Division of Nephrology and Hypertension, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | | |
Collapse
|
15
|
Ke B, Zhao Z, Ye X, Gao Z, Manganiello V, Wu B, Ye J. Inactivation of NF-κB p65 (RelA) in Liver Improves Insulin Sensitivity and Inhibits cAMP/PKA Pathway. Diabetes 2015; 64:3355-62. [PMID: 26038580 PMCID: PMC4587638 DOI: 10.2337/db15-0242] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 05/20/2015] [Indexed: 01/07/2023]
Abstract
The transcription factor nuclear factor-κB (NF-κB) mediates inflammation and stress signals in cells. To test NF-κB in the control of hepatic insulin sensitivity, we inactivated NF-κB in the livers of C57BL/6 mice through deletion of the p65 gene, which was achieved by crossing floxed-p65 and Alb-cre mice to generate L-p65-knockout (KO) mice. KO mice did not exhibit any alterations in growth, reproduction, and body weight while on a chow diet. However, the mice on a high-fat diet (HFD) exhibited an improvement in systemic insulin sensitivity. Hepatic insulin sensitivity was enhanced as indicated by increased pyruvate tolerance, Akt phosphorylation, and decreased gene expression in hepatic gluconeogenesis. In the liver, a decrease in intracellular cAMP was observed with decreased CREB phosphorylation. Cyclic nucleotide phosphodiesterase-3B (PDE3B), a cAMP-degrading enzyme, was increased in mRNA and protein as a result of the absence of NF-κB activity. NF-κB was found to inhibit PDE3B transcription through three DNA-binding sites in the gene promoter in response to tumor necrosis factor-α. Body composition, food intake, energy expenditure, and systemic and hepatic inflammation were not significantly altered in KO mice on HFD. These data suggest that NF-κB inhibits hepatic insulin sensitivity by upregulating cAMP through suppression of PDE3B gene transcription.
Collapse
Affiliation(s)
- Bilun Ke
- Department of Gastroenterology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| | - Zhiyun Zhao
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| | - Xin Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| | - Zhanguo Gao
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Vincent Manganiello
- Pulmonary Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Bin Wu
- Department of Gastroenterology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianping Ye
- Antioxidant and Gene Regulation Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA
| |
Collapse
|
16
|
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
|
17
|
Targeted disruption of PDE3B, but not PDE3A, protects murine heart from ischemia/reperfusion injury. Proc Natl Acad Sci U S A 2015; 112:E2253-62. [PMID: 25877153 DOI: 10.1073/pnas.1416230112] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although inhibition of cyclic nucleotide phosphodiesterase type 3 (PDE3) has been reported to protect rodent heart against ischemia/reperfusion (I/R) injury, neither the specific PDE3 isoform involved nor the underlying mechanisms have been identified. Targeted disruption of PDE3 subfamily B (PDE3B), but not of PDE3 subfamily A (PDE3A), protected mouse heart from I/R injury in vivo and in vitro, with reduced infarct size and improved cardiac function. The cardioprotective effect in PDE3B(-/-) heart was reversed by blocking cAMP-dependent PKA and by paxilline, an inhibitor of mitochondrial calcium-activated K channels, the opening of which is potentiated by cAMP/PKA signaling. Compared with WT mitochondria, PDE3B(-/-) mitochondria were enriched in antiapoptotic Bcl-2, produced less reactive oxygen species, and more frequently contacted transverse tubules where PDE3B was localized with caveolin-3. Moreover, a PDE3B(-/-) mitochondrial fraction containing connexin-43 and caveolin-3 was more resistant to Ca(2+)-induced opening of the mitochondrial permeability transition pore. Proteomics analyses indicated that PDE3B(-/-) heart mitochondria fractions were enriched in buoyant ischemia-induced caveolin-3-enriched fractions (ICEFs) containing cardioprotective proteins. Accumulation of proteins into ICEFs was PKA dependent and was achieved by ischemic preconditioning or treatment of WT heart with the PDE3 inhibitor cilostamide. Taken together, these findings indicate that PDE3B deletion confers cardioprotective effects because of cAMP/PKA-induced preconditioning, which is associated with the accumulation of proteins with cardioprotective function in ICEFs. To our knowledge, our study is the first to define a role for PDE3B in cardioprotection against I/R injury and suggests PDE3B as a target for cardiovascular therapies.
Collapse
|
18
|
Shao YX, Huang M, Cui W, Feng LJ, Wu Y, Cai Y, Li Z, Zhu X, Liu P, Wan Y, Ke H, Luo HB. Discovery of a phosphodiesterase 9A inhibitor as a potential hypoglycemic agent. J Med Chem 2014; 57:10304-13. [PMID: 25432025 PMCID: PMC4281101 DOI: 10.1021/jm500836h] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
Phosphodiesterase 9 (PDE9) inhibitors
have been studied as potential therapeutics for treatment of diabetes
and Alzheimer’s disease. Here we report a potent PDE9 inhibitor 3r that has an IC50 of 0.6 nM and >150-fold
selectivity over other PDEs. The HepG2 cell-based assay shows that 3r inhibits the mRNA expression of phosphoenolpyruvate carboxykinase
and glucose 6-phosphatase. These activities of 3r, together
with the reasonable pharmacokinetic properties and no acute toxicity
at 1200 mg/kg dosage, suggest its potential as a hypoglycemic agent.
The crystal structure of PDE9-3r reveals significantly
different conformation and hydrogen bonding pattern of 3r from those of previously published 28s. Both 3r and 28s form a hydrogen bond with Tyr424,
a unique PDE9 residue (except for PDE8), but 3r shows
an additional hydrogen bond with Ala452. This structure information
might be useful for design of PDE9 inhibitors.
Collapse
Affiliation(s)
- Yong-xian Shao
- School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou 510006, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ahmad F, Murata T, Shimizu K, Degerman E, Maurice D, Manganiello V. Cyclic nucleotide phosphodiesterases: important signaling modulators and therapeutic targets. Oral Dis 2014; 21:e25-50. [PMID: 25056711 DOI: 10.1111/odi.12275] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/09/2014] [Indexed: 02/06/2023]
Abstract
By catalyzing hydrolysis of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), cyclic nucleotide phosphodiesterases are critical regulators of their intracellular concentrations and their biological effects. As these intracellular second messengers control many cellular homeostatic processes, dysregulation of their signals and signaling pathways initiate or modulate pathophysiological pathways related to various disease states, including erectile dysfunction, pulmonary hypertension, acute refractory cardiac failure, intermittent claudication, chronic obstructive pulmonary disease, and psoriasis. Alterations in expression of PDEs and PDE-gene mutations (especially mutations in PDE6, PDE8B, PDE11A, and PDE4) have been implicated in various diseases and cancer pathologies. PDEs also play important role in formation and function of multimolecular signaling/regulatory complexes, called signalosomes. At specific intracellular locations, individual PDEs, together with pathway-specific signaling molecules, regulators, and effectors, are incorporated into specific signalosomes, where they facilitate and regulate compartmentalization of cyclic nucleotide signaling pathways and specific cellular functions. Currently, only a limited number of PDE inhibitors (PDE3, PDE4, PDE5 inhibitors) are used in clinical practice. Future paths to novel drug discovery include the crystal structure-based design approach, which has resulted in generation of more effective family-selective inhibitors, as well as burgeoning development of strategies to alter compartmentalized cyclic nucleotide signaling pathways by selectively targeting individual PDEs and their signalosome partners.
Collapse
Affiliation(s)
- F Ahmad
- Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
20
|
Maurice DH, Ke H, Ahmad F, Wang Y, Chung J, Manganiello VC. Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov 2014; 13:290-314. [PMID: 24687066 DOI: 10.1038/nrd4228] [Citation(s) in RCA: 595] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) catalyse the hydrolysis of cyclic AMP and cyclic GMP, thereby regulating the intracellular concentrations of these cyclic nucleotides, their signalling pathways and, consequently, myriad biological responses in health and disease. Currently, a small number of PDE inhibitors are used clinically for treating the pathophysiological dysregulation of cyclic nucleotide signalling in several disorders, including erectile dysfunction, pulmonary hypertension, acute refractory cardiac failure, intermittent claudication and chronic obstructive pulmonary disease. However, pharmaceutical interest in PDEs has been reignited by the increasing understanding of the roles of individual PDEs in regulating the subcellular compartmentalization of specific cyclic nucleotide signalling pathways, by the structure-based design of novel specific inhibitors and by the development of more sophisticated strategies to target individual PDE variants.
Collapse
Affiliation(s)
- Donald H Maurice
- Biomedical and Molecular Sciences, Queen's University, Kingston K7L3N6, Ontario, Canada
| | - Hengming Ke
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Faiyaz Ahmad
- Cardiovascular and Pulmonary Branch, The National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Yousheng Wang
- Beijing Technology and Business University, Beijing 100048, China
| | - Jay Chung
- Genetics and Developmental Biology Center, The National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Vincent C Manganiello
- Cardiovascular and Pulmonary Branch, The National Heart, Lung and Blood Institute, US National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|