1
|
Petty LE, Chen HH, Frankel EG, Zhu W, Downie CG, Graff M, Lin P, Sharma P, Zhang X, Scartozzi AC, Roshani R, Landman JM, Boehnke M, Bowden DW, Chambers JC, Mahajan A, McCarthy MI, Ng MCY, Sim X, Spracklen CN, Zhang W, Preuss M, Bottinger EP, Nadkarni GN, Loos RJF, Chen YDI, Tan J, Ipp E, Genter P, Emery LS, Louie T, Sofer T, Stilp AM, Taylor KD, Xiang AH, Buchanan TA, Roll K, Gao C, Palmer ND, Norris JM, Wagenknecht LE, Nousome D, Varma R, McKean-Cowdin R, Guo X, Hai Y, Hsueh W, Sandow K, Parra EJ, Cruz M, Valladares-Salgado A, Wacher-Rodarte N, Rotter JI, Goodarzi MO, Rich SS, Bertoni A, Raffel LJ, Nadler JL, Kandeel FR, Duggirala R, Blangero J, Lehman DM, DeFronzo RA, Thameem F, Wang Y, Gahagan S, Blanco E, Burrows R, Huerta-Chagoya A, Florez JC, Tusie-Luna T, González-Villalpando C, Orozco L, Haiman CA, Hanis CL, Rohde R, Whitsel EA, Reiner AP, Kooperberg C, Li Y, Duan Q, Lee M, Correa-Burrows P, Fried SK, North KE, McCormick JB, Fisher-Hoch SP, Gamazon ER, Morris AP, Mercader JM, Highland HM, Below JE. Large-scale multi-omics analyses in Hispanic/Latino populations identify genes for cardiometabolic traits. Nat Commun 2025; 16:3438. [PMID: 40210677 PMCID: PMC11985957 DOI: 10.1038/s41467-025-58574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 03/21/2025] [Indexed: 04/12/2025] Open
Abstract
Here, we present a multi-omics study of type 2 diabetes and quantitative blood lipid and lipoprotein traits conducted to date in Hispanic/Latino populations (nmax = 63,184). We conduct a meta-analysis of 16 type 2 diabetes and 19 lipid trait GWAS, identifying 20 genome-wide significant loci for type 2 diabetes, including one novel locus and novel signals at two known loci, based on fine-mapping. We also identify sixty-one genome-wide significant loci across the lipid/lipoprotein traits, including nine novel loci, and novel signals at 19 known loci through fine-mapping. Next, we analyze genetically regulated expression, perform Mendelian randomization, and analyze association with transcriptomic and proteomic measure using multi-omics data from a Hispanic/Latino population. Using this approach, we identify genes linked to type 2 diabetes and lipid/lipoprotein traits, including TMEM205 and NEDD9 for HDL cholesterol, TREH for triglycerides, and ANXA4 for type 2 diabetes.
Collapse
Affiliation(s)
- Lauren E Petty
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Elizabeth G Frankel
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wanying Zhu
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carolina G Downie
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Phillip Lin
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Priya Sharma
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alyssa C Scartozzi
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rashedeh Roshani
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua M Landman
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Donald W Bowden
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Anubha Mahajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Maggie C Y Ng
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology and Biostatistics, University of Massachusetts-Amherst, Amherst, MA, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Hasso-Plattner-Institut, Potsdam, Germany
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jingyi Tan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Eli Ipp
- Division of Endocrinology & Metabolism, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pauline Genter
- Division of Endocrinology & Metabolism, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leslie S Emery
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tamar Sofer
- Departments of Medicine and Biostatistics, Harvard University, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Adrienne M Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Anny H Xiang
- Department of Research & Evaluation, Division of Biostatistics Research, Kaiser Permanente of Southern California, Pasadena, CA, USA
| | - Thomas A Buchanan
- Department of Medicine, Division of Endocrine, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Kathryn Roll
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-, Salem, NC, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jill M Norris
- Department of Epidemiology, University of Colorado Denver, Aurora, CO, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Darryl Nousome
- Department of Preventative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Rohit Varma
- Southern California Eye Institute, CHA Hollywood Presbyterian Medical Center, Los Angeles, CA, USA
| | - Roberta McKean-Cowdin
- Department of Preventative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yang Hai
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Willa Hsueh
- Department of Internal Medicine, Diabetes and Metabolism Research Center, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kevin Sandow
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Esteban J Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, ON, Canada
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Adan Valladares-Salgado
- Unidad de Investigación Médica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Niels Wacher-Rodarte
- Unidad de Investigación Médica en Epidemiologia Clinica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mark O Goodarzi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Alain Bertoni
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Leslie J Raffel
- Department of Pediatrics, Division of Medical Genetics, UCI Irvine School of Medicine, Irvine, CA, USA
| | - Jerry L Nadler
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Fouad R Kandeel
- Departments of Clinical Diabetes, Endocrinology & Metabolism and Translational Research and Cellular Therapeutics, City of Hope, Duarte, CA, USA
| | | | - John Blangero
- Human Genetics and STDOI, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Donna M Lehman
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ralph A DeFronzo
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Farook Thameem
- Department of Biochemistry, Faculty of Medicine, Health Science Center, Kuwait University, Safat, Kuwait
| | - Yujie Wang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sheila Gahagan
- Division of Academic General Pediatrics, Child Development and Community Health, University of California at San Diego, San Diego, CA, USA
| | - Estela Blanco
- College y Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Raquel Burrows
- Department of Public Health Nutrition, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Alicia Huerta-Chagoya
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Tusie-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Clicerio González-Villalpando
- Centro de Estudios en Diabetes, Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Centro de Investigacion en Salud Poblacional, Instituto Nacional de Salud Pública, Mexico City, Mexico
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Craig L Hanis
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rebecca Rohde
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Yun Li
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Qing Duan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miryoung Lee
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Paulina Correa-Burrows
- Department of Public Health Nutrition, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Susan K Fried
- Diabetes, Obesity Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joseph B McCormick
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Clare Hall & MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
| | - Josep M Mercader
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
2
|
Gentile R, Feudi D, Sallicandro L, Biagini A. Can the Tumor Microenvironment Alter Ion Channels? Unraveling Their Role in Cancer. Cancers (Basel) 2025; 17:1244. [PMID: 40227837 PMCID: PMC11988140 DOI: 10.3390/cancers17071244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/29/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Neoplastic cells are characterized by metabolic reprogramming, known as the Warburg effect, in which glucose metabolism is predominantly directed toward aerobic glycolysis, with reduced mitochondrial oxidative phosphorylation and increased lactate production even in the presence of oxygen. This phenomenon provides cancer cells with a proliferative advantage, allowing them to rapidly produce energy (in the form of ATP) and generate metabolic intermediates necessary for the biosynthesis of macromolecules essential for cell growth. It is important to understand the role of ion channels in the tumor context since they participate in various physiological processes and in the regulation of the tumor microenvironment. These changes may contribute to the development and transformation of cancer cells, as well as affect the communication between cells and the surrounding microenvironment, including impaired or altered expression and functionality of ion channels. Therefore, the aim of this review is to elucidate the impact of the tumor microenvironment on the electrical properties of the cellular membranes in several cancers as a possible therapeutic target.
Collapse
Affiliation(s)
- Rosaria Gentile
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
| | - Davide Feudi
- Department of Biostatistics, Epidemiology and Public Health, University of Padua, Via L. Loredan 18, 35131 Padova, Italy;
| | - Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Biagini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| |
Collapse
|
3
|
York NW, Yan Z, Osipovich AB, Tate A, Patel S, Piston DW, Magnuson MA, Remedi MS, Nichols CG. Loss of β-Cell KATP Reduces Ca2+ Sensitivity of Insulin Secretion and Trpm5 Expression. Diabetes 2025; 74:376-383. [PMID: 39666394 PMCID: PMC11842610 DOI: 10.2337/db24-0650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
Loss-of-function mutations in ATP-sensitive potassium (KATP) channels cause hyperexcitability and insulin hypersecretion, resulting in congenital hyperinsulinism (CHI). Paradoxically, despite the initial insulin hypersecretion, many CHI cases, as well as KATP knockout (KO) animals, eventually "crossover" to undersecretion and even diabetes. Here, we confirm that Sur1 KO islets exhibit higher intracellular concentration of calcium ion ([Ca2+]i) at all concentrations of glucose but show decreased glucose-stimulated insulin secretion. However, when [Ca2+]i is artificially elevated by increasing extracellular [Ca2+], insulin secretion from Sur1 KO islets increases to the same levels as in wild-type (WT) islets. This indicates that a right-shift in [Ca2+]i dependence of insulin secretion, rather than loss of insulin content or intrinsic secretability, is the primary cause for the crossover. Chronic pharmacological inhibition of KATP channel activity by slow release of glibenclamide in pellet-implanted mice causes a very similar crossover to glucose intolerance and impaired insulin secretion seen in Sur1 KO animals. Whole-islet and single-cell transcriptomic analysis reveal markedly reduced Trpm5 in both conditions. Glibenclamide pellet-implanted Trpm5 KO mice also exhibited significant glucose intolerance. However, this was not as severe as in WT animals, which suggests decreased expression of Trpm5 may play a small role in the disruption of insulin secretion with KATP loss. ARTICLE HIGHLIGHTS Congenital hyperinsulinism caused by loss of ATP-sensitive potassium (KATP) channels crosses over to unexplained undersecretion. Why does loss of β-cell KATP channel activity result in undersecretion of insulin and glucose tolerance, despite elevated intracellular concentration of calcium ion ([Ca2+]i) levels? Superelevation of [Ca2+]i in supraphysiological extracellular [Ca2+] boosted secretion from Sur1 knockout (KO) islets to the same levels as WT, indicating a right-shift in [Ca2+]i dependence of secretion. Transcriptomic analysis revealed markedly reduced β-cell Trpm5 in the absence of KATP. KATP inhibition in Trpm5 KO mice still caused significant glucose intolerance, but slightly less severe than in WT animals. Right-shifted [Ca2+]i dependence of secretion explains crossover. Downregulation of Trpm5 may be involved.
Collapse
Affiliation(s)
- Nathaniel W. York
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Zihan Yan
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - Anna B. Osipovich
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Abbie Tate
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Sumit Patel
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - David W. Piston
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Mark A. Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Maria S. Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - Colin G. Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
4
|
Ives CM, Şahin AT, Thomson NJ, Zachariae U. A hydrophobic funnel governs monovalent cation selectivity in the ion channel TRPM5. Biophys J 2024; 123:3304-3316. [PMID: 39086136 PMCID: PMC11480762 DOI: 10.1016/j.bpj.2024.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
A key capability of ion channels is the facilitation of selective permeation of certain ionic species across cellular membranes at high rates. Due to their physiological significance, ion channels are of great pharmaceutical interest as drug targets. The polymodal signal-detecting transient receptor potential (TRP) superfamily of ion channels forms a particularly promising group of drug targets. While most members of this family permeate a broad range of cations including Ca2+, TRPM4 and TRPM5 are unique due to their strong monovalent selectivity and impermeability for divalent cations. Here, we investigated the mechanistic basis for their unique monovalent selectivity by in silico electrophysiology simulations of TRPM5. Our simulations reveal an unusual mechanism of cation selectivity, which is underpinned by the function of the central channel cavity alongside the selectivity filter. Our results suggest that a subtle hydrophobic barrier at the cavity entrance ("hydrophobic funnel") enables monovalent but not divalent cations to pass and occupy the cavity at physiologically relevant membrane voltages. Monovalent cations then permeate efficiently by a cooperative, distant knock-on mechanism between two binding regions in the extracellular pore vestibule and the central cavity. By contrast, divalent cations do not enter or interact favorably with the channel cavity due to its raised hydrophobicity. Hydrophilic mutations in the transition zone between the selectivity filter and the central channel cavity abolish the barrier for divalent cations, enabling both monovalent and divalent cations to traverse TRPM5.
Collapse
Affiliation(s)
- Callum M Ives
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Alp Tegin Şahin
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom; School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Neil J Thomson
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ulrich Zachariae
- Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom; Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
5
|
Yang B, Ma D, Zhu X, Wu Z, An Q, Zhao J, Gao X, Zhang L. Roles of TRP and PIEZO receptors in autoimmune diseases. Expert Rev Mol Med 2024; 26:e10. [PMID: 38659380 PMCID: PMC11140548 DOI: 10.1017/erm.2023.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/15/2023] [Accepted: 08/21/2023] [Indexed: 04/26/2024]
Abstract
Autoimmune diseases are pathological autoimmune reactions in the body caused by various factors, which can lead to tissue damage and organ dysfunction. They can be divided into organ-specific and systemic autoimmune diseases. These diseases usually involve various body systems, including the blood, muscles, bones, joints and soft tissues. The transient receptor potential (TRP) and PIEZO receptors, which resulted in David Julius and Ardem Patapoutian winning the Nobel Prize in Physiology or Medicine in 2021, attracted people's attention. Most current studies on TRP and PIEZO receptors in autoimmune diseases have been carried out on animal model, only few clinical studies have been conducted. Therefore, this study aimed to review existing studies on TRP and PIEZO to understand the roles of these receptors in autoimmune diseases, which may help elucidate novel treatment strategies.
Collapse
Affiliation(s)
- Baoqi Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xueqing Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Zewen Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
6
|
Wang KY, Wu SM, Yao ZJ, Zhu YX, Han X. Insufficient TRPM5 Mediates Lipotoxicity-induced Pancreatic β-cell Dysfunction. Curr Med Sci 2024; 44:346-354. [PMID: 38517672 DOI: 10.1007/s11596-023-2795-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 08/28/2023] [Indexed: 03/24/2024]
Abstract
OBJECTIVE While the reduction of transient receptor potential channel subfamily M member 5 (TRPM5) has been reported in islet cells from type 2 diabetic (T2D) mouse models, its role in lipotoxicity-induced pancreatic β-cell dysfunction remains unclear. This study aims to study its role. METHODS Pancreas slices were prepared from mice subjected to a high-fat-diet (HFD) at different time points, and TRPM5 expression in the pancreatic β cells was examined using immunofluorescence staining. Glucose-stimulated insulin secretion (GSIS) defects caused by lipotoxicity were mimicked by saturated fatty acid palmitate (Palm). Primary mouse islets and mouse insulinoma MIN6 cells were treated with Palm, and the TRPM5 expression was detected using qRT-PCR and Western blotting. Palm-induced GSIS defects were measured following siRNA-based Trpm5 knockdown. The detrimental effects of Palm on primary mouse islets were also assessed after overexpressing Trpm5 via an adenovirus-derived Trpm5 (Ad-Trpm5). RESULTS HFD feeding decreased the mRNA levels and protein expression of TRPM5 in mouse pancreatic islets. Palm reduced TRPM5 protein expression in a time- and dose-dependent manner in MIN6 cells. Palm also inhibited TRPM5 expression in primary mouse islets. Knockdown of Trpm5 inhibited insulin secretion upon high glucose stimulation but had little effect on insulin biosynthesis. Overexpression of Trpm5 reversed Palm-induced GSIS defects and the production of functional maturation molecules unique to β cells. CONCLUSION Our findings suggest that lipotoxicity inhibits TRPM5 expression in pancreatic β cells both in vivo and in vitro and, in turn, drives β-cell dysfunction.
Collapse
Affiliation(s)
- Kai-Yuan Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Shi-Mei Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Zheng-Jian Yao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Yun-Xia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
7
|
Nmarneh A, Priel A. TRPM5 activation depends on a synergistic effect of calcium and PKC phosphorylation. Commun Biol 2024; 7:369. [PMID: 38538847 PMCID: PMC10973328 DOI: 10.1038/s42003-024-06054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/15/2024] [Indexed: 12/14/2024] Open
Abstract
Transient receptor potential melastatin 5 (TRPM5) is a calcium-activated monovalent-specific ion channel involved in insulin secretion and taste transduction, making it an attractive target for drug development in various pathologies. While TRPM5 activation involves ligand binding to Gq/G-protein coupled receptors (GPCR) and subsequent elevation of intracellular calcium levels, recent reports suggest the need for additional molecular determinants. Hence, the mechanism of TRPM5 activation remains to be elucidated. Here, we show that PKC phosphorylation and the elevation of intracellular Ca2+ levels are required for TRPM5 activation, with PKC phosphorylation being crucial for channel-evoked currents, primarily at physiological membrane potentials. In contrast, physiological relevant calcium levels alone only induce TRPM5 activation at positive voltages. Our findings highlight the necessity of coordinated intracellular calcium release and PKC phosphorylation for TRPM5 activation. Thus, our results suggest that regulation of PKC activity could be a promising therapeutic target for diseases associated with TRPM5 modulation.
Collapse
Affiliation(s)
- Alaa Nmarneh
- The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Karem, Jerusalem, 9112102, Israel
| | - Avi Priel
- The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Karem, Jerusalem, 9112102, Israel.
| |
Collapse
|
8
|
Richter P, Andersen G, Kahlenberg K, Mueller AU, Pirkwieser P, Boger V, Somoza V. Sodium-Permeable Ion Channels TRPM4 and TRPM5 are Functional in Human Gastric Parietal Cells in Culture and Modulate the Cellular Response to Bitter-Tasting Food Constituents. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4906-4917. [PMID: 38378185 PMCID: PMC10921469 DOI: 10.1021/acs.jafc.3c09085] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/22/2024]
Abstract
Gastric parietal cells secrete chloride ions and protons to form hydrochloric acid. Besides endogenous stimulants, e.g., acetylcholine, bitter-tasting food constituents, e.g., caffeine, induce proton secretion via interaction with bitter taste receptors (TAS2Rs), leading to increased cytosolic Ca2+ and cAMP concentrations. We hypothesized TAS2R activation by bitter tastants to result in proton secretion via cellular Na+ influx mediated by transient receptor potential channels (TRP) M4 and M5 in immortalized human parietal HGT-1 cells. Using the food-derived TAS2R agonists caffeine and l-arginine, we demonstrate both bitter compounds to induce a TRPM4/M5-mediated Na+ influx, with EC50 values of 0.65 and 10.38 mM, respectively, that stimulates cellular proton secretion. Functional involvement of TAS2Rs in the caffeine-evoked effect was demonstrated by means of the TAS2R antagonist homoeriodictyol, and stably CRISPR-Cas9-edited TAS2R43ko cells. Building on previous results, these data further support the suitability of HGT-1 cells as a surrogate cell model for taste cells. In addition, TRPM4/M5 mediated a Na+ influx after stimulating HGT-1 cells with the acetylcholine analogue carbachol, indicating an interaction of the digestion-associated cholinergic pathway with a taste-signaling pathway in parietal cells.
Collapse
Affiliation(s)
- Phil Richter
- TUM
School of Life Sciences Weihenstephan, Technical
University of Munich, Alte Akademie 8, Freising 85354, Germany
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Str.
34, Freising 85354, Germany
| | - Gaby Andersen
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Str.
34, Freising 85354, Germany
| | - Kristin Kahlenberg
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Str.
34, Freising 85354, Germany
| | - Alina Ulrike Mueller
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Str.
34, Freising 85354, Germany
| | - Philip Pirkwieser
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Str.
34, Freising 85354, Germany
| | - Valerie Boger
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Str.
34, Freising 85354, Germany
| | - Veronika Somoza
- Leibniz
Institute for Food Systems Biology at the Technical University of
Munich, Lise-Meitner-Str.
34, Freising 85354, Germany
- Chair
of Nutritional Systems Biology, TUM School of Life Sciences, Technical University of Munich, Lise-Meitner-Str. 34, Freising 85354, Germany
- Department
of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), Vienna 1090, Austria
| |
Collapse
|
9
|
Zheng C, An T, Liang Z, Lv B, Liu Y, Hu X, Zhang Y, Liu N, Tao S, Deng R, Liu J, Jiang G. Revealing the mechanism of quinoa on type 2 diabetes based on intestinal flora and taste pathways. Food Sci Nutr 2023; 11:7930-7945. [PMID: 38107122 PMCID: PMC10724620 DOI: 10.1002/fsn3.3710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 09/03/2023] [Accepted: 09/08/2023] [Indexed: 12/19/2023] Open
Abstract
To investigate the antidiabetic effects and mechanisms of quinoa on type 2 diabetes mellitus (T2DM) mice model. In this context, we induced the T2DM mice model with a high-fat diet (HFD) combined with streptozotocin (STZ), followed by treatment with a quinoa diet. To explore the impact of quinoa on the intestinal flora, we predicted and validated its potential mechanism of hypoglycemic effect through network pharmacology, molecular docking, western blot, and immunohistochemistry (IHC). We found that quinoa could significantly improve abnormal glucolipid metabolism in T2DM mice. Further analysis showed that quinoa contributed to the improvement of gut microbiota composition positively. Moreover, it could downregulate the expression of TAS1R3 and TRPM5 in the colon. A total of 72 active components were identified by network pharmacology. Among them, TAS1R3 and TRPM5 were successfully docked with the core components of quinoa. These findings confirm that quinoa may exert hypoglycemic effects through gut microbiota and the TAS1R3/TRPM5 taste signaling pathway.
Collapse
Affiliation(s)
- Chun‐Yan Zheng
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| | - Tian An
- School of Traditional Chinese MedicineCapital Medical UniversityBeijingChina
| | - Zheng‐Ting Liang
- Traditional Chinese Medicine SchoolXinjiang Medical UniversityXinjiangChina
| | - Bo‐Han Lv
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| | - Yu‐Tong Liu
- Gansu Pure High‐Land Agricultural Science and Technology Limited CompanyLanzhouChina
- Zhong Li Science and Technology Limited CompanyBeijingChina
| | - Xue‐Hong Hu
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| | - Yue‐Lin Zhang
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| | - Nan‐Nan Liu
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| | - Si‐Yu Tao
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| | - Ru‐Xue Deng
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| | - Jia‐Xian Liu
- Gansu Pure High‐Land Agricultural Science and Technology Limited CompanyLanzhouChina
- Zhong Li Science and Technology Limited CompanyBeijingChina
| | - Guang‐Jian Jiang
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
10
|
Chulkov EG, Rohr CM, Marchant JS. Praziquantel activates a native cation current in Schistosoma mansoni. FRONTIERS IN PARASITOLOGY 2023; 2:1285177. [PMID: 39816816 PMCID: PMC11732042 DOI: 10.3389/fpara.2023.1285177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/26/2023] [Indexed: 01/18/2025]
Abstract
Introduction Praziquantel (PZQ), an anthelmintic drug discovered in the 1970s, is still used to treat schistosomiasis and various other infections caused by parasitic flatworms. PZQ causes a triad of phenotypic effects on schistosome worms - rapid depolarization, muscle contraction, and damage throughout the worm tegument. The molecular target mediating these effects has been intimated as a Ca2+-permeable ion channel, but native currents evoked by PZQ have not been reported in any schistosome cell type. The properties of the endogenous PZQ activated conductance therefore remain unknown. Methods Here, invasive electrophysiology was used to probe for responses to PZQ from different locales in a living schistosome worm. Results and discussion No direct response was seen in tegument-derived vesicles, or from the sub-tegumental muscle layer despite the presence of voltage-operated currents. However, PZQ rapidly triggered a sustained, non-selective cation current in recordings from neuronal tissue, targeting both the anterior ganglion and the main longitudinal nerve cord. The biophysical signature of this PZQ-evoked current resolved at single channel resolution matched that of a transient receptor potential ion channel named TRPMPZQ, recently proposed as the molecular target of PZQ. The endogenous PZQ-evoked current was also inhibited by a validated TRPMPZQ antagonist. PZQ therefore is a neuroactive anthelmintic, causing a sustained depolarization through ion channels with the characteristics of TRPMPZQ.
Collapse
Affiliation(s)
| | | | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
11
|
HU WEI, WARTMANN THOMAS, STRECKER MARCO, PERRAKIS ARISTOTELIS, CRONER ROLAND, SZALLASI ARPAD, SHI WENJIE, KAHLERT ULFD. Transient receptor potential channels as predictive marker and potential indicator of chemoresistance in colon cancer. Oncol Res 2023; 32:227-239. [PMID: 38188686 PMCID: PMC10767253 DOI: 10.32604/or.2023.043053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/13/2023] [Indexed: 01/09/2024] Open
Abstract
Transient receptor potential (TRP) channels are strongly associated with colon cancer development and progression. This study leveraged a multivariate Cox regression model on publicly available datasets to construct a TRP channels-associated gene signature, with further validation of signature in real world samples from our hospital treated patient samples. Kaplan-Meier (K-M) survival analysis and receiver operating characteristic (ROC) curves were employed to evaluate this gene signature's predictive accuracy and robustness in both training and testing cohorts, respectively. Additionally, the study utilized the CIBERSORT algorithm and single-sample gene set enrichment analysis to explore the signature's immune infiltration landscape and underlying functional implications. The support vector machine algorithm was applied to evaluate the signature's potential in predicting chemotherapy outcomes. The findings unveiled a novel three TRP channels-related gene signature (MCOLN1, TRPM5, and TRPV4) in colon adenocarcinoma (COAD). The ROC and K-M survival curves in the training dataset (AUC = 0.761; p = 1.58e-05) and testing dataset (AUC = 0.699; p = 0.004) showed the signature's robust predictive capability for the overall survival of COAD patients. Analysis of the immune infiltration landscape associated with the signature revealed higher immune infiltration, especially an increased presence of M2 macrophages, in high-risk group patients compared to their low-risk counterparts. High-risk score patients also exhibited potential responsiveness to immune checkpoint inhibitor therapy, evident through increased CD86 and PD-1 expression profiles. Moreover, the TRPM5 gene within the signature was highly expressed in the chemoresistance group (p = 0.00095) and associated with poor prognosis (p = 0.036) in COAD patients, highlighting its role as a hub gene of chemoresistance. Ultimately, this signature emerged as an independent prognosis factor for COAD patients (p = 6.48e-06) and expression of model gene are validated by public data and real-world patients. Overall, this bioinformatics study provides valuable insights into the prognostic implications and potential chemotherapy resistance mechanisms associated with TRPs-related genes in colon cancer.
Collapse
Affiliation(s)
- WEI HU
- The Fourth Clinical Medical College of Yangzhou University, Nantong Rich Hospital, Nantong, China
| | - THOMAS WARTMANN
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - MARCO STRECKER
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ARISTOTELIS PERRAKIS
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ROLAND CRONER
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ARPAD SZALLASI
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - WENJIE SHI
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ULF D. KAHLERT
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
12
|
Sakata N, Yoshimatsu G, Kawakami R, Aoyagi C, Kodama S. Optimal temperature for the long-term culture of adult porcine islets for xenotransplantation. Front Immunol 2023; 14:1280668. [PMID: 37901206 PMCID: PMC10611499 DOI: 10.3389/fimmu.2023.1280668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 10/31/2023] Open
Abstract
Porcine islet xenotransplantation represents a promising therapy for severe diabetes mellitus. Long-term culture of porcine islets is a crucial challenge to permit the on-demand provision of islets. We aimed to identify the optimal temperature for the long-term culture of adult porcine islets for xenotransplantation. We evaluated the factors potentially influencing successful 28-day culture of islets at 24°C and 37°C, and found that culture at 37°C contributed to the stability of the morphology of the islets, the proliferation of islet cells, and the recovery of endocrine function, indicated by the expression of genes involved in pancreatic development, hormone production, and glucose-stimulated insulin secretion. These advantages may be provided by islet-derived CD146-positive stellate cells. The efficacy of xenotransplantation using islets cultured for a long time at 37°C was similar to that of overnight-cultured islets. In conclusion, 37°C might be a suitable temperature for the long-term culture of porcine islets, but further modifications will be required for successful xenotransplantation in a clinical setting.
Collapse
Affiliation(s)
- Naoaki Sakata
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Ryo Kawakami
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Chikao Aoyagi
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Shohta Kodama
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| |
Collapse
|
13
|
Kõks S. Genomics of Wolfram Syndrome 1 (WFS1). Biomolecules 2023; 13:1346. [PMID: 37759745 PMCID: PMC10527379 DOI: 10.3390/biom13091346] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Wolfram Syndrome (WFS) is a rare, autosomal, recessive neurogenetic disorder that affects many organ systems. It is characterised by diabetes insipidus, diabetes mellites, optic atrophy, and deafness and, therefore, is also known as DIDMOAD. Nearly 15,000-30,000 people are affected by WFS worldwide, and, on average, patients suffering from WFS die at 30 years of age, usually from central respiratory failure caused by massive brain atrophy. The more prevalent of the two kinds of WFS is WFS1, which is a monogenic disease and caused by the loss of the WFS1 gene, whereas WFS2, which is more uncommon, is caused by mutations in the CISD2 gene. Currently, there is no treatment for WFS1 to increase the life expectancy of patients, and the treatments available do not significantly improve their quality of life. Understanding the genetics and the molecular mechanisms of WFS1 is essential to finding a cure. The inability of conventional medications to treat WFS1 points to the need for innovative strategies that must address the fundamental cause: the deletion of the WFS1 gene that leads to the profound ER stress and disturbances in proteostasis. An important approach here is to understand the mechanism of the cell degeneration after the deletion of the WFS1 gene and to describe the differences in these mechanisms for the different tissues. The studies so far have indicated that remarkable clinical heterogeneity is caused by the variable vulnerability caused by WFS1 mutations, and these differences cannot be attributed solely to the positions of mutations in the WFS1 gene. The present review gives a broader overview of the results from genomic studies on the WFS1 mouse model.
Collapse
Affiliation(s)
- Sulev Kõks
- Perron Institute for Neurological and Translational Science, 8 Verdun Street, Nedlands, WA 6009, Australia;
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
| |
Collapse
|
14
|
Yu Q, Gamayun I, Wartenberg P, Zhang Q, Qiao S, Kusumakshi S, Candlish S, Götz V, Wen S, Das D, Wyatt A, Wahl V, Ectors F, Kattler K, Yildiz D, Prevot V, Schwaninger M, Ternier G, Giacobini P, Ciofi P, Müller TD, Boehm U. Bitter taste cells in the ventricular walls of the murine brain regulate glucose homeostasis. Nat Commun 2023; 14:1588. [PMID: 36949050 PMCID: PMC10033832 DOI: 10.1038/s41467-023-37099-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/02/2023] [Indexed: 03/24/2023] Open
Abstract
The median eminence (ME) is a circumventricular organ at the base of the brain that controls body homeostasis. Tanycytes are its specialized glial cells that constitute the ventricular walls and regulate different physiological states, however individual signaling pathways in these cells are incompletely understood. Here, we identify a functional tanycyte subpopulation that expresses key taste transduction genes including bitter taste receptors, the G protein gustducin and the gustatory ion channel TRPM5 (M5). M5 tanycytes have access to blood-borne cues via processes extended towards diaphragmed endothelial fenestrations in the ME and mediate bidirectional communication between the cerebrospinal fluid and blood. This subpopulation responds to metabolic signals including leptin and other hormonal cues and is transcriptionally reprogrammed upon fasting. Acute M5 tanycyte activation induces insulin secretion and acute diphtheria toxin-mediated M5 tanycyte depletion results in impaired glucose tolerance in diet-induced obese mice. We provide a cellular and molecular framework that defines how bitter taste cells in the ME integrate chemosensation with metabolism.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Igor Gamayun
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Qian Zhang
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sen Qiao
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Soumya Kusumakshi
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Sarah Candlish
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Viktoria Götz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Shuping Wen
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Debajyoti Das
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Fabien Ectors
- FARAH Mammalian Transgenics Platform, Liège University, Liège, Belgium
| | - Kathrin Kattler
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Daniela Yildiz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Gaetan Ternier
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Philippe Ciofi
- Neurocentre Magendie - INSERM Unit 1215, University of Bordeaux, Bordeaux, France
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ulrich Boehm
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany.
| |
Collapse
|
15
|
Rached G, Saliba Y, Maddah D, Hajal J, Smayra V, Bakhos J, Groschner K, Birnbaumer L, Fares N. TRPC3 Regulates Islet Beta-Cell Insulin Secretion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204846. [PMID: 36642838 PMCID: PMC9951314 DOI: 10.1002/advs.202204846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/18/2022] [Indexed: 06/17/2023]
Abstract
Insulin release is tightly controlled by glucose-stimulated calcium (GSCa) through hitherto equivocal pathways. This study investigates TRPC3, a non-selective cation channel, as a critical regulator of insulin secretion and glucose control. TRPC3's involvement in glucose-stimulated insulin secretion (GSIS) is studied in human and animal islets. TRPC3-dependent in vivo insulin secretion is investigated using pharmacological tools and Trpc3-/- mice. TRPC3's involvement in islet glucose uptake and GSCa is explored using fluorescent glucose analogue 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-D-glucose and calcium imaging. TRPC3 modulation by a small-molecule activator, GSK1702934A, is evaluated in type 2 diabetic mice. TRPC3 is functionally expressed in human and mouse islet beta cells. TRPC3-controlled insulin secretion is KATP -independent and primarily mediated by diacylglycerol channel regulation of the cytosolic calcium oscillations following glucose stimulation. Conversely, glucose uptake in islets is independent of TRPC3. TRPC3 pharmacologic inhibition and knockout in mice lead to defective insulin secretion and glucose intolerance. Subsequently, TRPC3 activation through targeted small-molecule enhances insulin secretion and alleviates diabetes hallmarks in animals. This study imputes a function for TRPC3 at the onset of GSIS. These insights strengthen one's knowledge of insulin secretion physiology and set forth the TRPC3 channel as an appealing candidate for drug development in the treatment of diabetes.
Collapse
Affiliation(s)
- Gaëlle Rached
- Physiology and Pathophysiology Research LaboratoryPole of Technology and HealthFaculty of MedicineSaint Joseph University of BeirutPOBox. 17‐5208 ‐ Mar MikhaëlBeirut1104 2020Lebanon
| | - Youakim Saliba
- Physiology and Pathophysiology Research LaboratoryPole of Technology and HealthFaculty of MedicineSaint Joseph University of BeirutPOBox. 17‐5208 ‐ Mar MikhaëlBeirut1104 2020Lebanon
| | - Dina Maddah
- Physiology and Pathophysiology Research LaboratoryPole of Technology and HealthFaculty of MedicineSaint Joseph University of BeirutPOBox. 17‐5208 ‐ Mar MikhaëlBeirut1104 2020Lebanon
| | - Joelle Hajal
- Physiology and Pathophysiology Research LaboratoryPole of Technology and HealthFaculty of MedicineSaint Joseph University of BeirutPOBox. 17‐5208 ‐ Mar MikhaëlBeirut1104 2020Lebanon
| | - Viviane Smayra
- Faculty of MedicineSaint Joseph UniversitySaint Joseph University of BeirutPOBox. 17‐5208 ‐ Mar MikhaëlBeirut1104 2020Lebanon
| | - Jules‐Joel Bakhos
- Physiology and Pathophysiology Research LaboratoryPole of Technology and HealthFaculty of MedicineSaint Joseph University of BeirutPOBox. 17‐5208 ‐ Mar MikhaëlBeirut1104 2020Lebanon
| | - Klaus Groschner
- Gottfried‐Schatz‐Research‐Centre‐BiophysicsMedical University of GrazGraz8010Austria
| | - Lutz Birnbaumer
- School of Medical SciencesInstitute of Biomedical Research (BIOMED)Catholic University of ArgentinaBuenos AiresC1107AAZArgentina
- Signal Transduction LaboratoryNational Institute of Environmental Health SciencesResearch Triangle ParkDurhamNCC1107AAZUSA
| | - Nassim Fares
- Physiology and Pathophysiology Research LaboratoryPole of Technology and HealthFaculty of MedicineSaint Joseph University of BeirutPOBox. 17‐5208 ‐ Mar MikhaëlBeirut1104 2020Lebanon
| |
Collapse
|
16
|
Virginio C, Aldegheri L, Nola S, Brodbeck D, Brault L, Raveglia LF, Barilli A, Sabat M, Myers R. Identification of positive modulators of TRPM5 channel from a high-throughput screen using a fluorescent membrane potential assay. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:55-64. [PMID: 35058176 DOI: 10.1016/j.slasd.2021.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Transient Receptor Potential Melastatin 5 (TRPM5) is an intracellular calcium-activated cation-selective ion channel expressed in a variety of cell types. Dysfunction of this channel has recently been implied in a range of disease states including diabetes, enteric infections, inflammatory responses, parasitic infection and other pathologies. However, to date, agonists and positive modulators of this channel with sufficient selectivity to enable target validation studies have not been described, limiting the evaluation of TRPM5 biology and its potential as a drug target. We developed a high-throughput assay using a fluorescent membrane potential dye and a medium- and high-throughput electrophysiology assay using QPatch HTX and SyncroPatch 384PE. By employing these assays, we conducted a primary screening campaign and identified hit compounds as TRPM5 channel positive modulators. An initial selectivity profile confirmed hit selectivity to TRPM5 and is presented here. These small molecule TRPM5 compounds have a high potential both as early tool compounds to enable pharmacological studies of TRPM5 and as starting points for the development of potent, selective TRPM5 openers or positive modulators as novel drugs targeting several pathological states.
Collapse
Affiliation(s)
- Caterina Virginio
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy.
| | - Laura Aldegheri
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Selena Nola
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Daniela Brodbeck
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Laurent Brault
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Luca F Raveglia
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Alessio Barilli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Mark Sabat
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, CA 92121, United States
| | - Richard Myers
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, CA 92121, United States
| |
Collapse
|
17
|
López-Pérez A, Norlin S, Steneberg P, Remeseiro S, Edlund H, Hörnblad A. Pan-AMPK activator O304 prevents gene expression changes and remobilisation of histone marks in islets of diet-induced obese mice. Sci Rep 2021; 11:24410. [PMID: 34949756 PMCID: PMC8702551 DOI: 10.1038/s41598-021-03567-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/06/2021] [Indexed: 12/25/2022] Open
Abstract
AMP-activated protein kinase (AMPK) has an important role in cellular energy homeostasis and has emerged as a promising target for treatment of Type 2 Diabetes (T2D) due to its beneficial effects on insulin sensitivity and glucose homeostasis. O304 is a pan-AMPK activator that has been shown to improve glucose homeostasis in both mouse models of diabetes and in human T2D subjects. Here, we describe the genome-wide transcriptional profile and chromatin landscape of pancreatic islets following O304 treatment of mice fed high-fat diet (HFD). O304 largely prevented genome-wide gene expression changes associated with HFD feeding in CBA mice and these changes were associated with remodelling of active and repressive chromatin marks. In particular, the increased expression of the β-cell stress marker Aldh1a3 in islets from HFD-mice is completely abrogated following O304 treatment, which is accompanied by loss of active chromatin marks in the promoter as well as distant non-coding regions upstream of the Aldh1a3 gene. Moreover, O304 treatment restored dysfunctional glucose homeostasis as well as expression of key markers associated with β-cell function in mice with already established obesity. Our findings provide preclinical evidence that O304 is a promising therapeutic compound not only for T2D remission but also for restoration of β-cell function following remission of T2D diabetes.
Collapse
Affiliation(s)
- Ana López-Pérez
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures väg 12, 90187, Umeå, Sweden
| | - Stefan Norlin
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures väg 12, 90187, Umeå, Sweden
| | - Pär Steneberg
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures väg 12, 90187, Umeå, Sweden
| | - Silvia Remeseiro
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures väg 12, 90187, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, 90187, Umeå, Sweden
| | - Helena Edlund
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures väg 12, 90187, Umeå, Sweden.
| | - Andreas Hörnblad
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures väg 12, 90187, Umeå, Sweden.
| |
Collapse
|
18
|
Li X, Lai L, Su J, Chen S, Lin S, Wang B, Gao J, Zhang L, Yao K, Duan S. Novel association between a transient receptor potential cation channel subfamily M member 5 expression quantitative trait locus rs35197079 and decreased susceptibility of gestational diabetes mellitus in a Chinese population. J Diabetes Investig 2021; 12:2062-2070. [PMID: 33979016 PMCID: PMC8565411 DOI: 10.1111/jdi.13572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 12/29/2022] Open
Abstract
AIMS/INTRODUCTION Emerging evidence suggests that expression quantitative trait loci (eQTLs) are more likely to associate with complex diseases. Transient receptor potential cation channel subfamily M member 5 (TRPM5) is a ubiquitously expressed voltage-gated cation channel that acts indispensably to trigger insulin secretion in pancreatic β-cells. The present study evaluated the association between TRPM5 eQTL single-nucleotide polymorphisms and the risk of gestational diabetes mellitus (GDM) in a Chinese population. MATERIALS AND METHODS A total of 380 unrelated Chinese pregnant women including 241 GDM patients and 139 controls were included in this study. The eQTL single-nucleotide polymorphisms of TRPM5 were obtained from the GTEx eQTL Browser, and were subsequently genotyped using the Agena MassARRAY iPLEX platform. RESULTS Logistic regression analysis and linear regression analysis showed that rs35197079 and rs74848824 were significantly associated with reduced GDM risk and lower fasting plasma glucose levels after adjusting confounder factors in dominant genetic models. Stratification analysis based on pre-pregnancy body mass index validated a strong association between rs35197079 and GDM susceptibility in underweight and normal weight individuals. Luciferase and electrophoretic mobility shift assays carried out in rat pancreatic β-cells showed that rs35197079 was functional. CONCLUSIONS The TRPM5 eQTL single-nucleotide polymorphism rs35197079 was associated with decreased GDM susceptibility in a Chinese population, especially in underweight and normal weight pregnant women, and it was functional in modulating gene transcription.
Collapse
Affiliation(s)
- Xi Li
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Liping Lai
- Endocrine DepartmentFutian Center for Chronic Disease ControlShenzhenChina
| | - Jindi Su
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Shiguo Chen
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Sheng Lin
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Baojiang Wang
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Jian Gao
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Linghua Zhang
- Shenzhen Health Development Research CenterShenzhenChina
| | - Keqin Yao
- Shenzhen Health Development Research CenterShenzhenChina
| | - Shan Duan
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| |
Collapse
|
19
|
Zheng W, Li L, Li H. Phytochemicals modulate pancreatic islet β cell function through glucagon-like peptide-1-related mechanisms. Biochem Pharmacol 2021; 197:114817. [PMID: 34717897 DOI: 10.1016/j.bcp.2021.114817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/19/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) receptor-based therapies have been developed and extensively applied in clinical practice. GLP-1 plays an important role in improving glycemic homeostasis by stimulating insulin biosynthesis and secretion, suppressing glucagon activity, delaying gastric emptying, and reducing appetite and food ingestion. Furthermore, GLP-1 has positive effects on β-cell function by promoting β-cell proliferation and neogenesis while simultaneously reducing apoptosis. Here, we summarize possible mechanisms of action of GLP-1 upon pancreatic islets as well as describe phytochemicals that modulate pancreatic islet β cell function through glucagon-like peptide-1-related mechanisms. Together, this information provides potential lead compound candidates against diabetes that function as GLP-1 receptor-based pharmacotherapy.
Collapse
Affiliation(s)
- Wanfang Zheng
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Linghuan Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Hanbing Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; Section of Endocrinology, School of Medicine, Yale University, New Haven 06520, USA.
| |
Collapse
|
20
|
Ruan Z, Haley E, Orozco IJ, Sabat M, Myers R, Roth R, Du J, Lü W. Structures of the TRPM5 channel elucidate mechanisms of activation and inhibition. Nat Struct Mol Biol 2021; 28:604-613. [PMID: 34168372 PMCID: PMC8767786 DOI: 10.1038/s41594-021-00607-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023]
Abstract
The Ca2+-activated TRPM5 channel plays essential roles in taste perception and insulin secretion. However, the mechanism by which Ca2+ regulates TRPM5 activity remains elusive. We report cryo-EM structures of the zebrafish TRPM5 in an apo closed state, a Ca2+-bound open state, and an antagonist-bound inhibited state. We define two novel ligand binding sites: a Ca2+ site (CaICD) in the intracellular domain and an antagonist site in the transmembrane domain (TMD). The CaICD site is unique to TRPM5 and has two roles: modulating the voltage dependence and promoting Ca2+ binding to the CaTMD site, which is conserved throughout TRPM channels. Conformational changes initialized from both Ca2+ sites cooperatively open the ion-conducting pore. The antagonist NDNA wedges into the space between the S1-S4 domain and pore domain, stabilizing the transmembrane domain in an apo-like closed state. Our results lay the foundation for understanding the voltage-dependent TRPM channels and developing new therapeutic agents.
Collapse
Affiliation(s)
- Zheng Ruan
- Van Andel Institute, 333 Bostwick Ave., N.E., Grand Rapids, MI 49503
| | - Emery Haley
- Van Andel Institute, 333 Bostwick Ave., N.E., Grand Rapids, MI 49503
| | - Ian J. Orozco
- Van Andel Institute, 333 Bostwick Ave., N.E., Grand Rapids, MI 49503
| | - Mark Sabat
- Takeda California Inc, 9625 Towne Centre Dr, San Diego, CA 92121
| | - Richard Myers
- Takeda California Inc, 9625 Towne Centre Dr, San Diego, CA 92121
| | - Rebecca Roth
- Van Andel Institute, 333 Bostwick Ave., N.E., Grand Rapids, MI 49503
| | - Juan Du
- Van Andel Institute, 333 Bostwick Ave., N.E., Grand Rapids, MI 49503,CORRESPONDING AUTHOR: Correspondence and requests for materials should be addressed to J. D. () TEL: (616) 234-5358, FAX: 616-234-5170 or W. L. (). TEL: (616) 234-5022, FAX: 616-234-5170
| | - Wei Lü
- Van Andel Institute, 333 Bostwick Ave., N.E., Grand Rapids, MI 49503,CORRESPONDING AUTHOR: Correspondence and requests for materials should be addressed to J. D. () TEL: (616) 234-5358, FAX: 616-234-5170 or W. L. (). TEL: (616) 234-5022, FAX: 616-234-5170
| |
Collapse
|
21
|
Shi R, Fu Y, Zhao D, Boczek T, Wang W, Guo F. Cell death modulation by transient receptor potential melastatin channels TRPM2 and TRPM7 and their underlying molecular mechanisms. Biochem Pharmacol 2021; 190:114664. [PMID: 34175300 DOI: 10.1016/j.bcp.2021.114664] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
Transient receptor potential melastatin (TRPM) channels are members of the transient receptor potential (TRP) channels, a family of evolutionarily conserved integral membrane proteins. TRPM channels are nonselective cation channels, mediating the influx of various ions including Ca2+, Na+ and Zn2+. The function of TRPM channels is vital for cell proliferation, cell development and cell death. Cell death is a key procedure during embryonic development, organism homeostasis, aging and disease. The category of cell death modalities, beyond the traditionally defined concepts of necrosis, autophagy, and apoptosis, were extended with the discovery of pyroptosis, necroptosis and ferroptosis. As upstream signaling regulators of cell death, TRPM channels have been involved inrelevant pathologies. In this review, we introduced several cell death modalities, then summarized the contribution of TRPM channels (especially TRPM2 and TRPM7) to different cell death modalities and discussed the underlying regulatory mechanisms. Our work highlighted the possibility of TRPM channels as potential therapeutic targets in cell death-related diseases.
Collapse
Affiliation(s)
- Ruixue Shi
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yu Fu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Dongyi Zhao
- The University of Tokyo, Department of Pharmaceutical Science, 1130033, Japan
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, 92215, Poland.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
22
|
Glucose and NAADP trigger elementary intracellular β-cell Ca 2+ signals. Sci Rep 2021; 11:10714. [PMID: 34021189 PMCID: PMC8140081 DOI: 10.1038/s41598-021-88906-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/15/2021] [Indexed: 11/12/2022] Open
Abstract
Pancreatic β-cells release insulin upon a rise in blood glucose. The precise mechanisms of stimulus-secretion coupling, and its failure in Diabetes Mellitus Type 2, remain to be elucidated. The consensus model, as well as a class of currently prescribed anti-diabetic drugs, are based around the observation that glucose-evoked ATP production in β-cells leads to closure of cell membrane ATP-gated potassium (KATP) channels, plasma membrane depolarisation, Ca2+ influx, and finally the exocytosis of insulin granules. However, it has been demonstrated by the inactivation of this pathway using genetic and pharmacological means that closure of the KATP channel alone may not be sufficient to explain all β-cell responses to glucose elevation. We have previously proposed that NAADP-evoked Ca2+ release is an important step in stimulus-secretion coupling in pancreatic β-cells. Here we show using total internal reflection fluorescence (TIRF) microscopy that glucose as well as the Ca2+ mobilising messenger nicotinic acid adenine dinucleotide phosphate (NAADP), known to operate in β-cells, lead to highly localised elementary intracellular Ca2+ signals. These were found to be obscured by measurements of global Ca2+ signals and the action of powerful SERCA-based sequestration mechanisms at the endoplasmic reticulum (ER). Building on our previous work demonstrating that NAADP-evoked Ca2+ release is an important step in stimulus-secretion coupling in pancreatic β-cells, we provide here the first demonstration of elementary Ca2+ signals in response to NAADP, whose occurrence was previously suspected. Optical quantal analysis of these events reveals a unitary event amplitude equivalent to that of known elementary Ca2+ signalling events, inositol trisphosphate (IP3) receptor mediated blips, and ryanodine receptor mediated quarks. We propose that a mechanism based on these highly localised intracellular Ca2+ signalling events mediated by NAADP may initially operate in β-cells when they respond to elevations in blood glucose.
Collapse
|
23
|
Barilli A, Aldegheri L, Bianchi F, Brault L, Brodbeck D, Castelletti L, Feriani A, Lingard I, Myers R, Nola S, Piccoli L, Pompilio D, Raveglia LF, Salvagno C, Tassini S, Virginio C, Sabat M. From High-Throughput Screening to Target Validation: Benzo[ d]isothiazoles as Potent and Selective Agonists of Human Transient Receptor Potential Cation Channel Subfamily M Member 5 Possessing In Vivo Gastrointestinal Prokinetic Activity in Rodents. J Med Chem 2021; 64:5931-5955. [PMID: 33890770 DOI: 10.1021/acs.jmedchem.1c00065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transient receptor potential cation channel subfamily M member 5 (TRPM5) is a nonselective monovalent cation channel activated by intracellular Ca2+ increase. Within the gastrointestinal system, TRPM5 is expressed in the stoma, small intestine, and colon. In the search for a selective agonist of TRPM5 possessing in vivo gastrointestinal prokinetic activity, a high-throughput screening was performed and compound 1 was identified as a promising hit. Hit validation and hit to lead activities led to the discovery of a series of benzo[d]isothiazole derivatives. Among these, compounds 61 and 64 showed nanomolar activity and excellent selectivity (>100-fold) versus related cation channels. The in vivo drug metabolism and pharmacokinetic profile of compound 64 was found to be ideal for a compound acting locally at the intestinal level, with minimal absorption into systemic circulation. Compound 64 was tested in vivo in a mouse motility assay at 100 mg/kg, and demonstrated increased prokinetic activity.
Collapse
Affiliation(s)
- Alessio Barilli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laura Aldegheri
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Federica Bianchi
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laurent Brault
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Daniela Brodbeck
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laura Castelletti
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Aldo Feriani
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Iain Lingard
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Richard Myers
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Selena Nola
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Laura Piccoli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Daniela Pompilio
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Luca F Raveglia
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Cristian Salvagno
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Sabrina Tassini
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Caterina Virginio
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, Verona 37135, Italy
| | - Mark Sabat
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| |
Collapse
|
24
|
Abstract
Already for centuries, humankind is driven to understand the physiological and pathological mechanisms that occur in our brains. Today, we know that ion channels play an essential role in the regulation of neural processes and control many functions of the central nervous system. Ion channels present a diverse group of membrane-spanning proteins that allow ions to penetrate the insulating cell membrane upon opening of their channel pores. This regulated ion permeation results in different electrical and chemical signals that are necessary to maintain physiological excitatory and inhibitory processes in the brain. Therefore, it is no surprise that disturbances in the functions of cerebral ion channels can result in a plethora of neurological disorders, which present a tremendous health care burden for our current society. The identification of ion channel-related brain disorders also fuel the research into the roles of ion channel proteins in various brain states. In the last decade, mounting evidence has been collected that indicates a pivotal role for transient receptor potential (TRP) ion channels in the development and various physiological functions of the central nervous system. For instance, TRP channels modulate neurite growth, synaptic plasticity and integration, and are required for neuronal survival. Moreover, TRP channels are involved in numerous neurological disorders. TRPM3 belongs to the melastatin subfamily of TRP channels and represents a non-selective cation channel that can be activated by several different stimuli, including the neurosteroid pregnenolone sulfate, osmotic pressures and heat. The channel is best known as a peripheral nociceptive ion channel that participates in heat sensation. However, recent research identifies TRPM3 as an emerging new player in the brain. In this review, we summarize the available data regarding the roles of TRPM3 in the brain, and correlate these data with the neuropathological processes in which this ion channel may be involved.
Collapse
Affiliation(s)
- Katharina Held
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine and VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
| | - Balázs István Tóth
- Laboratory of Cellular and Molecular Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
25
|
Jimenez I, Prado Y, Marchant F, Otero C, Eltit F, Cabello-Verrugio C, Cerda O, Simon F. TRPM Channels in Human Diseases. Cells 2020; 9:E2604. [PMID: 33291725 PMCID: PMC7761947 DOI: 10.3390/cells9122604] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
The transient receptor potential melastatin (TRPM) subfamily belongs to the TRP cation channels family. Since the first cloning of TRPM1 in 1989, tremendous progress has been made in identifying novel members of the TRPM subfamily and their functions. The TRPM subfamily is composed of eight members consisting of four six-transmembrane domain subunits, resulting in homomeric or heteromeric channels. From a structural point of view, based on the homology sequence of the coiled-coil in the C-terminus, the eight TRPM members are clustered into four groups: TRPM1/M3, M2/M8, M4/M5 and M6/M7. TRPM subfamily members have been involved in several physiological functions. However, they are also linked to diverse pathophysiological human processes. Alterations in the expression and function of TRPM subfamily ion channels might generate several human diseases including cardiovascular and neurodegenerative alterations, organ dysfunction, cancer and many other channelopathies. These effects position them as remarkable putative targets for novel diagnostic strategies, drug design and therapeutic approaches. Here, we review the current knowledge about the main characteristics of all members of the TRPM family, focusing on their actions in human diseases.
Collapse
Affiliation(s)
- Ivanka Jimenez
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
| | - Yolanda Prado
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
| | - Felipe Marchant
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
| | - Carolina Otero
- Faculty of Medicine, School of Chemistry and Pharmacy, Universidad Andrés Bello, Santiago 8370186, Chile;
| | - Felipe Eltit
- Vancouver Prostate Centre, Vancouver, BC V6Z 1Y6, Canada;
- Department of Urological Sciences, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Claudio Cabello-Verrugio
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 7560484, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
| | - Oscar Cerda
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Felipe Simon
- Faculty of Life Science, Universidad Andrés Bello, Santiago 8370186, Chile; (I.J.); (Y.P.); (F.M.); (C.C.-V.)
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8380453, Chile;
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
| |
Collapse
|
26
|
Altered Transcription Factor Binding and Gene Bivalency in Islets of Intrauterine Growth Retarded Rats. Cells 2020; 9:cells9061435. [PMID: 32527043 PMCID: PMC7348746 DOI: 10.3390/cells9061435] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/30/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
Intrauterine growth retardation (IUGR), which induces epigenetic modifications and permanent changes in gene expression, has been associated with the development of type 2 diabetes. Using a rat model of IUGR, we performed ChIP-Seq to identify and map genome-wide histone modifications and gene dysregulation in islets from 2- and 10-week rats. IUGR induced significant changes in the enrichment of H3K4me3, H3K27me3, and H3K27Ac marks in both 2-wk and 10-wk islets, which were correlated with expression changes of multiple genes critical for islet function in IUGR islets. ChIP-Seq analysis showed that IUGR-induced histone mark changes were enriched at critical transcription factor binding motifs, such as C/EBPs, Ets1, Bcl6, Thrb, Ebf1, Sox9, and Mitf. These transcription factors were also identified as top upstream regulators in our previously published transcriptome study. In addition, our ChIP-seq data revealed more than 1000 potential bivalent genes as identified by enrichment of both H3K4me3 and H3K27me3. The poised state of many potential bivalent genes was altered by IUGR, particularly Acod1, Fgf21, Serpina11, Cdh16, Lrrc27, and Lrrc66, key islet genes. Collectively, our findings suggest alterations of histone modification in key transcription factors and genes that may contribute to long-term gene dysregulation and an abnormal islet phenotype in IUGR rats.
Collapse
|
27
|
Hanschmann EM, Petry SF, Eitner S, Maresch CC, Lingwal N, Lillig CH, Linn T. Paracrine regulation and improvement of β-cell function by thioredoxin. Redox Biol 2020; 34:101570. [PMID: 32473461 PMCID: PMC7260591 DOI: 10.1016/j.redox.2020.101570] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
The failure of insulin-producing β-cells is the underlying cause of hyperglycemia in diabetes mellitus. β-cell decay has been linked to hypoxia, chronic inflammation, and oxidative stress. Thioredoxin (Trx) proteins are major actors in redox signaling and essential for signal transduction and the cellular stress response. We have analyzed the cytosolic, mitochondrial, and extracellular Trx system proteins in hypoxic and cytokine-induced stress using β-cell culture, isolated pancreatic islets, and pancreatic islet transplantation modelling low oxygen supply. Protein levels of cytosolic Trx1 and Trx reductase (TrxR) 1 significantly decreased, while mitochondrial Trx2 and TrxR2 increased upon hypoxia and reoxygenation. Interestingly, Trx1 was secreted by β-cells during hypoxia. Moreover, murine and human pancreatic islet grafts released Trx1 upon glucose stimulation. Survival of transplanted islets was substantially impaired by the TrxR inhibitor auranofin. Since a release was prominent upon hypoxia, putative paracrine effects of Trx1 on β-cells were examined. In fact, exogenously added recombinant hTrx1 mitigated apoptosis and preserved glucose sensitivity in pancreatic islets subjected to hypoxia and inflammatory stimuli, dependent on its redox activity. Human subjects were studied, demonstrating a transient increase in extracellular Trx1 in serum after glucose challenge. This increase correlated with better pancreatic islet function. Moreover, hTrx1 inhibited the migration of primary murine macrophages. In conclusion, our study offers evidence for paracrine functions of extracellular Trx1 that improve the survival and function of pancreatic β-cells.
Collapse
Affiliation(s)
- Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, Germany; Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Susanne Eitner
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, Germany
| | | | - Neelam Lingwal
- Clinical Research Unit, Center of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, Germany.
| | - Thomas Linn
- Clinical Research Unit, Center of Internal Medicine, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
28
|
Yip L, Fuhlbrigge R, Alkhataybeh R, Fathman CG. Gene Expression Analysis of the Pre-Diabetic Pancreas to Identify Pathogenic Mechanisms and Biomarkers of Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:609271. [PMID: 33424774 PMCID: PMC7793767 DOI: 10.3389/fendo.2020.609271] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/16/2020] [Indexed: 12/28/2022] Open
Abstract
Type 1 Diabetes (T1D) occurs as a result of the autoimmune destruction of pancreatic β-cells by self-reactive T cells. The etiology of this disease is complex and difficult to study due to a lack of disease-relevant tissues from pre-diabetic individuals. In this study, we performed gene expression analysis on human pancreas tissues obtained from the Network of Pancreatic Organ Donors with Diabetes (nPOD), and showed that 155 genes were differentially expressed by ≥2-fold in the pancreata of autoantibody-positive (AA+) at-risk individuals compared to healthy controls. Only 48 of these genes remained changed by ≥2-fold in the pancreata of established T1D patients. Pathway analysis of these genes showed a significant association with various immune pathways. We were able to validate the differential expression of eight disease-relevant genes by QPCR analysis: A significant upregulation of CADM2, and downregulation of TRPM5, CRH, PDK4, ANGPL4, CLEC4D, RSG16, and FCGR2B was confirmed in the pancreata of AA+ individuals versus controls. Studies have already implicated FCGR2B in the pathogenesis of disease in non-obese diabetic (NOD) mice. Here we showed that CADM2, TRPM5, PDK4, and ANGPL4 were similarly changed in the pancreata of pre-diabetic 12-week-old NOD mice compared to NOD.B10 controls, suggesting a possible role for these genes in the pathogenesis of both T1D and NOD disease. The loss of the leukocyte-specific gene, FCGR2B, in the pancreata of AA+ individuals, is particularly interesting, as it may serve as a potential whole blood biomarker of disease progression. To test this, we quantified FCGR2B expression in peripheral blood samples of T1D patients, and AA+ and AA- first-degree relatives of T1D patients enrolled in the TrialNet Pathway to Prevention study. We showed that FCGR2B was significantly reduced in the peripheral blood of AA+ individuals compared to AA- controls. Together, these findings demonstrate that gene expression analysis of pancreatic tissue and peripheral blood samples can be used to identify disease-relevant genes and pathways and potential biomarkers of disease progression in T1D.
Collapse
|
29
|
Sarmiento BE, Santos Menezes LF, Schwartz EF. Insulin Release Mechanism Modulated by Toxins Isolated from Animal Venoms: From Basic Research to Drug Development Prospects. Molecules 2019; 24:E1846. [PMID: 31091684 PMCID: PMC6571724 DOI: 10.3390/molecules24101846] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/23/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
Venom from mammals, amphibians, snakes, arachnids, sea anemones and insects provides diverse sources of peptides with different potential medical applications. Several of these peptides have already been converted into drugs and some are still in the clinical phase. Diabetes type 2 is one of the diseases with the highest mortality rate worldwide, requiring specific attention. Diverse drugs are available (e.g., Sulfonylureas) for effective treatment, but with several adverse secondary effects, most of them related to the low specificity of these compounds to the target. In this context, the search for specific and high-affinity compounds for the management of this metabolic disease is growing. Toxins isolated from animal venom have high specificity and affinity for different molecular targets, of which the most important are ion channels. This review will present an overview about the electrical activity of the ion channels present in pancreatic β cells that are involved in the insulin secretion process, in addition to the diversity of peptides that can interact and modulate the electrical activity of pancreatic β cells. The importance of prospecting bioactive peptides for therapeutic use is also reinforced.
Collapse
Affiliation(s)
- Beatriz Elena Sarmiento
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil.
| | - Luis Felipe Santos Menezes
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil.
| | - Elisabeth F Schwartz
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil.
| |
Collapse
|
30
|
Farashi S, Sasanpour P, Rafii-Tabar H. The role of the transient receptor potential melastatin5 (TRPM5) channels in the pancreatic β-cell electrical activity: A computational modeling study. Comput Biol Chem 2018; 76:101-108. [DOI: 10.1016/j.compbiolchem.2018.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 03/31/2018] [Accepted: 05/15/2018] [Indexed: 01/27/2023]
|
31
|
Computational modeling of the effect of temperature variations on human pancreatic β-cell activity. J Therm Biol 2018; 75:69-80. [PMID: 30017054 DOI: 10.1016/j.jtherbio.2018.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 11/22/2022]
Abstract
The effect of temperature variations on the pancreatic β-cell activity and the role of different model compartments in temperature sensing have been investigated using a computational modeling approach. The results of our study show that temperature variations by several degrees can change the dynamical states of the β-cell system. In addition, temperature variations can alter the characteristic features of the membrane voltage, which correlates with insulin secretion. Simulation results show that the ion channels such as the L-type calcium, the hERG potassium, sodium channels and the glycolysis pathway are the possible sites for sensing temperature variation. Results indicate that for a small temperature change, even though the frequency and amplitude of electrical activity are altered, the area under the membrane potential curve remains almost unchanged, which implies the existence of a thermoregulatory mechanism for preserving the amount of insulin secretion. Furthermore, the computational analysis shows that the β-cell electrical activity exhibits a bursting pattern in physiological temperature (37 °C) while in vitro studies reported almost the spiking activity at lower temperatures. Since hormone-secreting systems work more efficient in bursting mode, we propose that the pancreatic β-cell works better in the physiological temperature compared with the reference temperature (33 °C).
Collapse
|
32
|
Vennekens R, Mesuere M, Philippaert K. TRPM5 in the battle against diabetes and obesity. Acta Physiol (Oxf) 2018; 222. [PMID: 28834354 DOI: 10.1111/apha.12949] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/06/2017] [Accepted: 08/09/2017] [Indexed: 12/22/2022]
Abstract
TRPM5 is a non-selective monovalent cation channel activated by increases in intracellular Ca2+ . It has a distinct expression pattern: expression is detected in chemosensitive tissues from solitary chemosensory cells to the taste receptor cells and in pancreatic β-cells. The role of TRPM5 has been investigated with the use of knockout mouse models. Trpm5-/- mice have a lack of type II taste perception and show reduced glucose-induced insulin secretion. Expression levels of TRPM5 are reduced in obese, leptin-signalling-deficient mice, and mutations in TRPM5 have been associated with type II diabetes and metabolic syndrome. In this review, we aim to give an overview of the activation, selectivity, modulation and physiological roles of TRPM5.
Collapse
Affiliation(s)
- R. Vennekens
- VIB Center for Brain & Disease Research; Leuven Belgium
- Laboratory of Ion Channel Research; TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine; KU Leuven, Leuven Belgium
| | - M. Mesuere
- VIB Center for Brain & Disease Research; Leuven Belgium
- Laboratory of Ion Channel Research; TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine; KU Leuven, Leuven Belgium
| | - K. Philippaert
- VIB Center for Brain & Disease Research; Leuven Belgium
- Laboratory of Ion Channel Research; TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine; KU Leuven, Leuven Belgium
| |
Collapse
|
33
|
Maeda T, Suzuki A, Koga K, Miyamoto C, Maehata Y, Ozawa S, Hata RI, Nagashima Y, Nabeshima K, Miyazaki K, Kato Y. TRPM5 mediates acidic extracellular pH signaling and TRPM5 inhibition reduces spontaneous metastasis in mouse B16-BL6 melanoma cells. Oncotarget 2017; 8:78312-78326. [PMID: 29108231 PMCID: PMC5667964 DOI: 10.18632/oncotarget.20826] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/27/2017] [Indexed: 01/13/2023] Open
Abstract
Extracellular acidity is a hallmark of solid tumors and is associated with metastasis in the tumor microenvironment. Acidic extracellular pH (pH e ) has been found to increase intracellular Ca2+ and matrix metalloproteinase-9 (MMP-9) expression by activating NF-κB in the mouse B16 melanoma model. The present study assessed whether TRPM5, an intracellular Ca2+-dependent monovalent cation channel, is associated with acidic pH e signaling and induction of MMP-9 expression in this mouse melanoma model. Treatment of B16 cells with Trpm5 siRNA reduced acidic pH e -induced MMP-9 expression. Enforced expression of Trpm5 increased the rate of acidic pH e -induced MMP-9 expression, as well as increasing experimental lung metastasis. This genetic manipulation did not alter the pH e critical for MMP-9 induction but simply amplified the percentage of inducible MMP-9 at each pH e . Treatment of tumor bearing mice with triphenylphosphine oxide (TPPO), an inhibitor of TRPM5, significantly reduced spontaneous lung metastasis. In silico analysis of clinical samples showed that high TRPM5 mRNA expression correlated with poor overall survival rate in patients with melanoma and gastric cancer but not in patients with cancers of the ovary, lung, breast, and rectum. These results showed that TRPM5 amplifies acidic pH e signaling and may be a promising target for preventing metastasis of some types of tumor.
Collapse
Affiliation(s)
- Toyonobu Maeda
- Department of Oral Function and Molecular Biology, Ohu University School of Dentistry, Koriyama 963-8611, Japan
| | - Atsuko Suzuki
- Department of Oral Function and Molecular Biology, Ohu University School of Dentistry, Koriyama 963-8611, Japan
| | - Kaori Koga
- Department of Pathology, Fukuoka University School of Medicine and Hospital, Fukuoka 814-0180, Japan
| | - Chihiro Miyamoto
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka 238-8580, Japan
| | - Yojiro Maehata
- Department of Oral Science, Kanagawa Dental University Graduate School of Dentistry, Yokosuka 238-8580, Japan
| | - Shigeyuki Ozawa
- Department of Dentomaxillofacial Diagnosis and Treatment, Kanagawa Dental University Graduate School of Dentistry, Yokosuka 238-8580, Japan
| | - Ryu-Ichiro Hata
- Department of Dentomaxillofacial Diagnosis and Treatment, Kanagawa Dental University Graduate School of Dentistry, Yokosuka 238-8580, Japan
- Oral Health Science Research Center, Kanagawa Dental University Graduate School of Dentistry, Yokosuka 238-8580, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women’s Medical University Hospital, Tokyo 162-8666, Japan
| | - Kazuki Nabeshima
- Department of Pathology, Fukuoka University School of Medicine and Hospital, Fukuoka 814-0180, Japan
| | - Kaoru Miyazaki
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan
| | - Yasumasa Kato
- Department of Oral Function and Molecular Biology, Ohu University School of Dentistry, Koriyama 963-8611, Japan
| |
Collapse
|
34
|
Actions and Regulation of Ionotropic Cannabinoid Receptors. ADVANCES IN PHARMACOLOGY 2017; 80:249-289. [PMID: 28826537 DOI: 10.1016/bs.apha.2017.04.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Almost three decades have passed since the identification of the two specific metabotropic receptors mediating cannabinoid pharmacology. Thereafter, many cannabinoid effects, both at central and peripheral levels, have been well documented and characterized. However, numerous evidences demonstrated that these pharmacological actions could not be attributable solely to the activation of CB1 and CB2 receptors since several important cannabimimetic actions have been found in biological systems lacking CB1 or CB2 gene such as in specific cell lines or transgenic mice. It is now well accepted that, beyond their receptor-mediated effects, these molecules can act also via CB1/CB2-receptor-independent mechanism. Cannabinoids have been demonstrated to modulate several voltage-gated channels (including Ca2+, Na+, and various type of K+ channels), ligand-gated ion channels (i.e., GABA, glycine), and ion-transporting membranes proteins such as transient potential receptor class (TRP) channels. The first direct, cannabinoid receptor-independent interaction was reported on the function of serotonin 5-HT3 receptor-ion channel complex. Similar effects were reported also on the other above mentioned ion channels. In the early ninety, studies searching for endogenous modulators of L-type Ca2+ channels identified anandamide as ligand for L-type Ca2+ channel. Later investigations indicated that other types of Ca2+ currents are also affected by endocannabinoids, and, in the late ninety, it was discovered that endocannabinoids activate the vanilloid receptor subtype 1 (TRPV1), and nowadays, it is known that (endo)cannabinoids gate at least five distinct TRP channels. This chapter focuses on cannabinoid regulation of ion channels and lays special emphasis on their action at transient receptor channels.
Collapse
|
35
|
N-acyl Taurines and Acylcarnitines Cause an Imbalance in Insulin Synthesis and Secretion Provoking β Cell Dysfunction in Type 2 Diabetes. Cell Metab 2017; 25:1334-1347.e4. [PMID: 28591636 DOI: 10.1016/j.cmet.2017.04.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/14/2017] [Accepted: 04/13/2017] [Indexed: 02/06/2023]
Abstract
The processes contributing to β cell dysfunction in type 2 diabetes (T2D) are uncertain, largely because it is difficult to access β cells in their intact immediate environment. We examined the pathophysiology of β cells under T2D progression directly in pancreatic tissues. We used MALDI imaging of Langerhans islets (LHIs) within mouse tissues or from human tissues to generate in situ-omics data, which we supported with in vitro experiments. Molecular interaction networks provided information on functional pathways and molecules. We found that stearoylcarnitine accumulated in β cells, leading to arrest of insulin synthesis and energy deficiency via excessive β-oxidation and depletion of TCA cycle and oxidative phosphorylation metabolites. Acetylcarnitine and an accumulation of N-acyl taurines, a group not previously detected in β cells, provoked insulin secretion. Thus, β cell dysfunction results from enhanced insulin secretion combined with an arrest of insulin synthesis.
Collapse
|
36
|
Ciardo MG, Ferrer-Montiel A. Lipids as central modulators of sensory TRP channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1615-1628. [PMID: 28432033 DOI: 10.1016/j.bbamem.2017.04.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/13/2017] [Accepted: 04/15/2017] [Indexed: 12/13/2022]
Abstract
The transient receptor potential (TRP) ion channel family is involved in a diversity of physiological processes including sensory and homeostatic functions, as well as muscle contraction and vasomotor control. Their dysfunction contributes to the etiology of several diseases, being validated as therapeutic targets. These ion channels may be activated by physical or chemical stimuli and their function is highly influenced by signaling molecules activated by extracellular signals. Notably, as integral membrane proteins, lipid molecules also modulate their membrane location and function either by direct interaction with the channel structure or by modulating the physico-chemical properties of the cellular membrane. This lipid-based modulatory effect is being considered an alternative and promising approach to regulate TRP channel dysfunction in diseases. Here, we review the current progress in this exciting field highlighting a complex channel regulation by a large diversity of lipid molecules and suggesting some diseases that may benefit from a membrane lipid therapy. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Av. De la Universidad s/n, Elche, Spain.
| |
Collapse
|
37
|
Steviol glycosides enhance pancreatic beta-cell function and taste sensation by potentiation of TRPM5 channel activity. Nat Commun 2017; 8:14733. [PMID: 28361903 PMCID: PMC5380970 DOI: 10.1038/ncomms14733] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/26/2017] [Indexed: 02/07/2023] Open
Abstract
Steviol glycosides (SGs), such as stevioside and rebaudioside A, are natural, non-caloric sweet-tasting organic molecules, present in extracts of the scrub plant Stevia rebaudiana, which are widely used as sweeteners in consumer foods and beverages. TRPM5 is a Ca2+-activated cation channel expressed in type II taste receptor cells and pancreatic β-cells. Here we show that stevioside, rebaudioside A and their aglycon steviol potentiate the activity of TRPM5. We find that SGs potentiate perception of bitter, sweet and umami taste, and enhance glucose-induced insulin secretion in a Trpm5-dependent manner. Daily consumption of stevioside prevents development of high-fat-diet-induced diabetic hyperglycaemia in wild-type mice, but not in Trpm5-/- mice. These results elucidate a molecular mechanism of action of SGs and identify TRPM5 as a potential target to prevent and treat type 2 diabetes.
Collapse
|
38
|
Trpm5 expression in the olfactory epithelium. Mol Cell Neurosci 2017; 80:75-88. [PMID: 28188885 DOI: 10.1016/j.mcn.2017.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 11/20/2022] Open
Abstract
The Ca2+-activated monovalent cation channel Trpm5 is a key element in chemotransduction of taste receptor cells of the tongue, but the extent to which Trpm5 channels are expressed in olfactory sensory neurons (OSNs) of the main olfactory epithelium (MOE) of adult mice as part of a specific pheromonal detection system is debated. Here, we used a novel Trpm5-IRES-Cre knockin strain to drive Cre recombinase expression, employed previously validated Trpm5 antibodies, performed in situ hybridization experiments to localize Trpm5 RNA, and searched extensively for Trpm5 splice variants in genetically-labeled, Trpm5-expressing MOE cells. In contrast to previous reports, we find no evidence for the existence in adult mouse OSNs of the classical Trpm5 channel known from taste cells. We show that Trpm5-expressing adult OSNs express a novel Trpm5 splice variant, Trpm5-9, that is unlikely to form a functional cation channel by itself. We also demonstrate that Trpm5 is transiently expressed in a subpopulation of mature OSNs in the embryonic olfactory epithelium, indicating that Trpm5 channels could play a specific role in utero during a narrow developmental time window. Ca2+ imaging with GCaMP3 under the control of the Trpm5-IRES-Cre allele using a newly developed MOE wholemount preparation of the adult olfactory epithelium reveals that Trpm5-GCaMP3 OSNs comprise a heterogeneous group of sensory neurons many of which can detect general odorants. Together, these studies are essential for understanding the role of transient receptor potential channels in mammalian olfaction.
Collapse
|
39
|
An optimized protocol for purification of functional islets of Langerhans. J Transl Med 2017; 97:70-83. [PMID: 27892930 DOI: 10.1038/labinvest.2016.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/21/2016] [Indexed: 12/31/2022] Open
Abstract
Islets of Langerhans and β-cell isolation constitute routinely used cell models for diabetic research, and refining islet isolation protocols and cell quality assessment is a high priority. Numerous protocols have been published describing isolate of islets, but often rigorous and systematic assessment of their integrity is lacking. Herein, we propose a new protocol for optimal generation of islets. Pancreases from mice and rats were excised and digested using a low-activity collagenase solution and islets were then purified by a series of sedimentations and a Percoll gradient. Islets were maintained in culture for 5 days, during which viability, pro/antiapoptotic, and islet-specific genes, glucose-stimulated calcium entry, glucose uptake, and insulin secretion were assessed. The commonly used islet isolation technique by collagenase injection through the common bile duct (CBD) was also performed and compared with the present approach. This new protocol produced islets that retained a healthy status as demonstrated by the yield of stable living cells. Furthermore, calcium oscillation, glucose uptake, and insulin secretion remained intact in the islet cultures. This was reproducible when many rodent species were used, and neither sex nor age affected the cells behavior. When compared with the CBD technique, islet physiology was similar. Finally, this approach was used to uncover new ion channel candidates implicated in insulin secretion. In conclusion, this study outlines an efficient protocol for islet preparation that may support research into new therapeutic targets in diabetes research.
Collapse
|
40
|
Ivask M, Hugill A, Kõks S. RNA-sequencing of WFS1-deficient pancreatic islets. Physiol Rep 2016; 4:4/7/e12750. [PMID: 27053292 PMCID: PMC4831324 DOI: 10.14814/phy2.12750] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 03/03/2016] [Indexed: 11/24/2022] Open
Abstract
Wolfram syndrome, an autosomal recessive disorder characterized by juvenile‐onset diabetes mellitus and optic atrophy, is caused by mutations in the WFS1 gene. WFS1 encodes an endoplasmic reticulum resident transmembrane protein. The Wfs1‐null mice exhibit progressive insulin deficiency and diabetes. The aim of this study was to describe the insulin secretion and transcriptome of pancreatic islets in WFS1‐deficient mice. WFS1‐deficient (Wfs1KO) mice had considerably less pancreatic islets than heterozygous (Wfs1HZ) or wild‐type (WT) mice. Wfs1KO pancreatic islets secreted less insulin after incubation in 2 and 10 mmol/L glucose and with tolbutamide solution compared to WT and Wfs1HZ islets, but not after stimulation with 20 mmol/L glucose. Differences in proinsulin amount were not statistically significant although there was a trend that Wfs1KO had an increased level of proinsulin. After incubation in 2 mmol/L glucose solution the proinsulin/insulin ratio in Wfs1KO was significantly higher than that of WT and Wfs1HZ. RNA‐seq from pancreatic islets found melastatin‐related transient receptor potential subfamily member 5 protein gene (Trpm5) to be downregulated in WFS1‐deficient mice. Functional annotation of RNA sequencing results showed that WFS1 deficiency influenced significantly the pathways related to tissue morphology, endocrine system development and function, molecular transport network.
Collapse
Affiliation(s)
- Marilin Ivask
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine University of Tartu, Tartu, Estonia
| | - Alison Hugill
- Mammalian Genetics Unit, Medical Research Council, Harwell, Oxfordshire, United Kingdom
| | - Sulev Kõks
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine University of Tartu, Tartu, Estonia
| |
Collapse
|
41
|
Ushiama S, Ishimaru Y, Narukawa M, Yoshioka M, Kozuka C, Watanabe N, Tsunoda M, Osakabe N, Asakura T, Masuzaki H, Abe K. Catecholamines Facilitate Fuel Expenditure and Protect Against Obesity via a Novel Network of the Gut-Brain Axis in Transcription Factor Skn-1-deficient Mice. EBioMedicine 2016; 8:60-71. [PMID: 27428419 PMCID: PMC4919597 DOI: 10.1016/j.ebiom.2016.04.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 11/26/2022] Open
Abstract
Taste signals and nutrient stimuli sensed by the gastrointestinal tract are transmitted to the brain to regulate feeding behavior and energy homeostasis. This system is referred to as the gut-brain axis. Here we show that both brush cells and type II taste cells are eliminated in the gastrointestinal tract of transcription factor Skn-1 knockout (KO) mice. Despite unaltered food intake, Skn-1 KO mice have reduced body weight with lower body fat due to increased energy expenditure. In this model, 24-h urinary excretion of catecholamines was significantly elevated, accompanied by increased fatty acid β-oxidation and fuel dissipation in skeletal muscle and impaired insulin secretion driven by glucose. These results suggest the existence of brain-mediated energy homeostatic pathways originating from brush cells and type II taste cells in the gastrointestinal tract and ending in peripheral tissues, including the adrenal glands. The discovery of food-derived factors that regulate these cells may open new avenues the treatment of obesity and diabetes. RESEARCH CONTEXT Taste signals and nutrient stimuli sensed by the gastrointestinal tract are transmitted to the brain to regulate feeding behavior and energy homeostasis along the gut-brain axis. We propose the concept that taste-receiving cells in the oral cavity and/or food-borne chemicals-receiving brush cells in the gut are involved in regulation of the body weight and adiposity via the brain. The discovery of food-derived factors that regulate these cells may open new avenues for the treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Shota Ushiama
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Yoshiro Ishimaru
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | - Masataka Narukawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Misako Yoshioka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Chisayo Kozuka
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan
| | - Naoki Watanabe
- Department of Bio-science and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Minuma-ku, Saitama 337-8570, Japan
| | - Makoto Tsunoda
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naomi Osakabe
- Department of Bio-science and Engineering, Shibaura Institute of Technology, 307 Fukasaku, Minuma-ku, Saitama 337-8570, Japan
| | - Tomiko Asakura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Hiroaki Masuzaki
- Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan
| | - Keiko Abe
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; Kanagawa Academy of Science and Technology, Takatsu-ku, Kawasaki-shi, Kanagawa, Japan.
| |
Collapse
|
42
|
Calcium Entry Through Thermosensory Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:265-304. [PMID: 27161233 DOI: 10.1007/978-3-319-26974-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ThermoTRPs are unique channels that mediate Na(+) and Ca(2+) currents in response to changes in ambient temperature. In combination with their activation by other physical and chemical stimuli, they are considered key integrators of environmental cues into neuronal excitability. Furthermore, roles of thermoTRPs in non-neuronal tissues are currently emerging such as insulin secretion in pancreatic β-cells, and links to cancer. Calcium permeability through thermoTRPs appears a central hallmark for their physiological and pathological activities. Moreover, it is currently being proposed that beyond working as a second messenger, Ca(2+) can function locally by acting on protein complexes near the membrane. Interestingly, thermoTRPs can enhance and expand the inherent plasticity of signalplexes by conferring them temperature, pH and lipid regulation through Ca(2+) signalling. Thus, unveiling the local role of Ca(2+) fluxes induced by thermoTRPs on the dynamics of membrane-attached signalling complexes as well as their significance in cellular processes, are central issues that will expand the opportunities for therapeutic intervention in disorders involving dysfunction of thermoTRP channels.
Collapse
|
43
|
Ablation of TRPM5 in Mice Results in Reduced Body Weight Gain and Improved Glucose Tolerance and Protects from Excessive Consumption of Sweet Palatable Food when Fed High Caloric Diets. PLoS One 2015; 10:e0138373. [PMID: 26397098 PMCID: PMC4580452 DOI: 10.1371/journal.pone.0138373] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 08/27/2015] [Indexed: 01/09/2023] Open
Abstract
The calcium activated cation channel transient receptor potential channel type M5 (TRPM5) is part of the downstream machinery of the taste receptors and have been shown to play a central role in taste signalling. In addition it is also found in other types of chemosensory cells in various parts of the body as well as in pancreatic β-cells. The aim of this study was to investigate the effects of TRPM5 gene ablation on body weight, insulin sensitivity and other metabolic parameters in long-term high caloric diet induced obesity. Trpm5-/- mice gained significantly less body weight and fat mass on both palatable carbohydrate and fat rich cafeteria diet and 60% high fat diet (HFD) and developed less insulin resistance compared to wild type mice. A main finding was the clearly improved glucose tolerance in Trpm5-/- mice compared to wild type mice on cafeteria diet, which was independent of body weight. In addition, it was shown that Trpm5-/- mice consumed the same amount of calories when fed a HFD only or a HFD in combination with a palatable chocolate ball, which is in contrast to wild type mice that increased their caloric intake when fed the combination, mainly due to excessive consumption of the chocolate ball. Thus the palatable sugar containing diet induced overeating was prevented in Trpm5-/- mice. This indicates that sweet taste induced overeating may be a cause for the increased energy intake and glucose intolerance development seen for wild type mice on a sugar and high fat rich cafeteria diet compared to when on a high fat diet. This study point to an important role for the taste signalling system and TRPM5 in diet induced glucose intolerance.
Collapse
|
44
|
Arredouani A, Ruas M, Collins SC, Parkesh R, Clough F, Pillinger T, Coltart G, Rietdorf K, Royle A, Johnson P, Braun M, Zhang Q, Sones W, Shimomura K, Morgan AJ, Lewis AM, Chuang KT, Tunn R, Gadea J, Teboul L, Heister PM, Tynan PW, Bellomo EA, Rutter GA, Rorsman P, Churchill GC, Parrington J, Galione A. Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP) and Endolysosomal Two-pore Channels Modulate Membrane Excitability and Stimulus-Secretion Coupling in Mouse Pancreatic β Cells. J Biol Chem 2015; 290:21376-21392. [PMID: 26152717 PMCID: PMC4571866 DOI: 10.1074/jbc.m115.671248] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/03/2015] [Indexed: 12/02/2022] Open
Abstract
Pancreatic β cells are electrically excitable and respond to elevated glucose concentrations with bursts of Ca(2+) action potentials due to the activation of voltage-dependent Ca(2+) channels (VDCCs), which leads to the exocytosis of insulin granules. We have examined the possible role of nicotinic acid adenine dinucleotide phosphate (NAADP)-mediated Ca(2+) release from intracellular stores during stimulus-secretion coupling in primary mouse pancreatic β cells. NAADP-regulated Ca(2+) release channels, likely two-pore channels (TPCs), have recently been shown to be a major mechanism for mobilizing Ca(2+) from the endolysosomal system, resulting in localized Ca(2+) signals. We show here that NAADP-mediated Ca(2+) release from endolysosomal Ca(2+) stores activates inward membrane currents and depolarizes the β cell to the threshold for VDCC activation and thereby contributes to glucose-evoked depolarization of the membrane potential during stimulus-response coupling. Selective pharmacological inhibition of NAADP-evoked Ca(2+) release or genetic ablation of endolysosomal TPC1 or TPC2 channels attenuates glucose- and sulfonylurea-induced membrane currents, depolarization, cytoplasmic Ca(2+) signals, and insulin secretion. Our findings implicate NAADP-evoked Ca(2+) release from acidic Ca(2+) storage organelles in stimulus-secretion coupling in β cells.
Collapse
Affiliation(s)
- Abdelilah Arredouani
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom,
| | - Margarida Ruas
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Stephan C Collins
- the Centre des Sciences du Gout et de l'Alimentation, Equipe 5, 9E Boulevard Jeanne d'Arc 21000 Dijon, France
| | - Raman Parkesh
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Frederick Clough
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Toby Pillinger
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - George Coltart
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Katja Rietdorf
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Andrew Royle
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Paul Johnson
- the Nuffield Department of Surgery, John Radcliffe Hospital, Headley Way, Headington, Oxford OX3 9DU, United Kingdom
| | - Matthias Braun
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - Quan Zhang
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - William Sones
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - Kenju Shimomura
- the Henry Wellcome Centre for Gene Function, Department of Physiology, Anatomy, and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, United Kingdom
| | - Anthony J Morgan
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Alexander M Lewis
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Kai-Ting Chuang
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Ruth Tunn
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Joaquin Gadea
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Lydia Teboul
- The Mary Lyon Centre, Medical Research Council Harwell, Oxfordshire OX11 0RD, United Kingdom
| | - Paula M Heister
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Patricia W Tynan
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Elisa A Bellomo
- the Centre des Sciences du Gout et de l'Alimentation, Equipe 5, 9E Boulevard Jeanne d'Arc 21000 Dijon, France
| | - Guy A Rutter
- the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Medicine, Imperial College London, Hammersmith Hospital, du Cane Road, London W12 0NN, United Kingdom, and
| | - Patrik Rorsman
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - Grant C Churchill
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - John Parrington
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom,
| | - Antony Galione
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom,
| |
Collapse
|
45
|
Smani T, Shapovalov G, Skryma R, Prevarskaya N, Rosado JA. Functional and physiopathological implications of TRP channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1772-82. [DOI: 10.1016/j.bbamcr.2015.04.016] [Citation(s) in RCA: 289] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/22/2015] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
|
46
|
Hiramoto M, Udagawa H, Watanabe A, Miyazawa K, Ishibashi N, Kawaguchi M, Uebanso T, Nishimura W, Nammo T, Yasuda K. Comparative analysis of type 2 diabetes-associated SNP alleles identifies allele-specific DNA-binding proteins for the KCNQ1 locus. Int J Mol Med 2015; 36:222-30. [PMID: 25955334 DOI: 10.3892/ijmm.2015.2203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/30/2015] [Indexed: 11/05/2022] Open
Abstract
Although recent genome-wide association studies (GWAS) have been extremely successful, it remains a big challenge to functionally annotate disease‑associated single nucleotide polymorphisms (SNPs), as the majority of these SNPs are located in non‑coding regions of the genome. In this study, we described a novel strategy for identifying the proteins that bind to the SNP‑containing locus in an allele‑specific manner and successfully applied this method to SNPs in the type 2 diabetes mellitus susceptibility gene, potassium voltage‑gated channel, KQT‑like subfamily Q, member 1 (KCNQ1). DNA fragments encompassing SNPs, and risk or non‑risk alleles were immobilized onto the novel nanobeads and DNA‑binding proteins were purified from the nuclear extracts of pancreatic β cells using these DNA‑immobilized nanobeads. Comparative analysis of the allele-specific DNA-binding proteins indicated that the affinities of several proteins for the examined SNPs differed between the alleles. Nuclear transcription factor Y (NF‑Y) specifically bound the non‑risk allele of the SNP rs2074196 region and stimulated the transcriptional activity of an artificial promoter containing SNP rs2074196 in an allele‑specific manner. These results suggest that SNP rs2074196 modulates the affinity of the locus for NF‑Y and possibly induces subsequent changes in gene expression. The findings of this study indicate that our comparative method using novel nanobeads is effective for the identification of allele‑specific DNA‑binding proteins, which may provide important clues for the functional impact of disease‑associated non‑coding SNPs.
Collapse
Affiliation(s)
- Masaki Hiramoto
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| | - Haruhide Udagawa
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| | - Atsushi Watanabe
- Laboratory of Research Advancement, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474‑8511, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Naoko Ishibashi
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| | - Miho Kawaguchi
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| | - Takashi Uebanso
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| | - Wataru Nishimura
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| | - Takao Nammo
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| | - Kazuki Yasuda
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo 162‑8655, Japan
| |
Collapse
|
47
|
Kusumakshi S, Voigt A, Hübner S, Hermans-Borgmeyer I, Ortalli A, Pyrski M, Dörr J, Zufall F, Flockerzi V, Meyerhof W, Montmayeur JP, Boehm U. A Binary Genetic Approach to Characterize TRPM5 Cells in Mice. Chem Senses 2015; 40:413-25. [PMID: 25940069 DOI: 10.1093/chemse/bjv023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transient receptor potential channel subfamily M member 5 (TRPM5) is an important downstream signaling component in a subset of taste receptor cells making it a potential target for taste modulation. Interestingly, TRPM5 has been detected in extra-oral tissues; however, the function of extra-gustatory TRPM5-expressing cells is less well understood. To facilitate visualization and manipulation of TRPM5-expressing cells in mice, we generated a Cre knock-in TRPM5 allele by homologous recombination. We then used the novel TRPM5-IRES-Cre mouse strain to report TRPM5 expression by activating a τGFP transgene. To confirm faithful coexpression of τGFP and TRPM5 we generated and validated a new anti-TRPM5 antiserum enabling us to analyze acute TRPM5 protein expression. τGFP cells were found in taste bud cells of the vallate, foliate, and fungiform papillae as well as in the palate. We also detected TRPM5 expression in several other tissues such as in the septal organ of Masera. Interestingly, in the olfactory epithelium of adult mice acute TRPM5 expression was detected in only one (short microvillar cells) of two cell populations previously reported to express TRPM5. The TRPM5-IC mouse strain described here represents a novel genetic tool and will facilitate the study and tissue-specific manipulation of TRPM5-expressing cells in vivo.
Collapse
Affiliation(s)
- Soumya Kusumakshi
- Department of Pharmacology and Toxicology, University of Saarland, School of Medicine, 66421 Homburg, Germany
| | - Anja Voigt
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Sandra Hübner
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Animals Service Group, UKE, Center for Molecular Neurobiology Hamburg (ZMNH), Falkenried 94, 20251 Hamburg, Germany
| | - Ana Ortalli
- Department of Pharmacology and Toxicology, University of Saarland, School of Medicine, 66421 Homburg, Germany, Centre des Sciences du Goût et de l'Alimentation, UMR6265 CNRS, UMR1324 INRA, Université de Bourgogne, 9E Boulevard Jeanne D'Arc, 21000 Dijon, France and
| | - Martina Pyrski
- Department of Physiology, University of Saarland, School of Medicine, 66421 Homburg, Germany
| | - Janka Dörr
- Department of Pharmacology and Toxicology, University of Saarland, School of Medicine, 66421 Homburg, Germany
| | - Frank Zufall
- Department of Physiology, University of Saarland, School of Medicine, 66421 Homburg, Germany
| | - Veit Flockerzi
- Department of Pharmacology and Toxicology, University of Saarland, School of Medicine, 66421 Homburg, Germany
| | - Wolfgang Meyerhof
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Jean-Pierre Montmayeur
- Centre des Sciences du Goût et de l'Alimentation, UMR6265 CNRS, UMR1324 INRA, Université de Bourgogne, 9E Boulevard Jeanne D'Arc, 21000 Dijon, France and
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, University of Saarland, School of Medicine, 66421 Homburg, Germany,
| |
Collapse
|
48
|
Yang SN, Shi Y, Yang G, Li Y, Yu J, Berggren PO. Ionic mechanisms in pancreatic β cell signaling. Cell Mol Life Sci 2014; 71:4149-77. [PMID: 25052376 PMCID: PMC11113777 DOI: 10.1007/s00018-014-1680-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 07/03/2014] [Accepted: 07/10/2014] [Indexed: 01/07/2023]
Abstract
The function and survival of pancreatic β cells critically rely on complex electrical signaling systems composed of a series of ionic events, namely fluxes of K(+), Na(+), Ca(2+) and Cl(-) across the β cell membranes. These electrical signaling systems not only sense events occurring in the extracellular space and intracellular milieu of pancreatic islet cells, but also control different β cell activities, most notably glucose-stimulated insulin secretion. Three major ion fluxes including K(+) efflux through ATP-sensitive K(+) (KATP) channels, the voltage-gated Ca(2+) (CaV) channel-mediated Ca(2+) influx and K(+) efflux through voltage-gated K(+) (KV) channels operate in the β cell. These ion fluxes set the resting membrane potential and the shape, rate and pattern of firing of action potentials under different metabolic conditions. The KATP channel-mediated K(+) efflux determines the resting membrane potential and keeps the excitability of the β cell at low levels. Ca(2+) influx through CaV1 channels, a major type of β cell CaV channels, causes the upstroke or depolarization phase of the action potential and regulates a wide range of β cell functions including the most elementary β cell function, insulin secretion. K(+) efflux mediated by KV2.1 delayed rectifier K(+) channels, a predominant form of β cell KV channels, brings about the downstroke or repolarization phase of the action potential, which acts as a brake for insulin secretion owing to shutting down the CaV channel-mediated Ca(2+) entry. These three ion channel-mediated ion fluxes are the most important ionic events in β cell signaling. This review concisely discusses various ionic mechanisms in β cell signaling and highlights KATP channel-, CaV1 channel- and KV2.1 channel-mediated ion fluxes.
Collapse
Affiliation(s)
- Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76, Stockholm, Sweden,
| | | | | | | | | | | |
Collapse
|
49
|
Nilius B, Szallasi A. Transient receptor potential channels as drug targets: from the science of basic research to the art of medicine. Pharmacol Rev 2014; 66:676-814. [PMID: 24951385 DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The large Trp gene family encodes transient receptor potential (TRP) proteins that form novel cation-selective ion channels. In mammals, 28 Trp channel genes have been identified. TRP proteins exhibit diverse permeation and gating properties and are involved in a plethora of physiologic functions with a strong impact on cellular sensing and signaling pathways. Indeed, mutations in human genes encoding TRP channels, the so-called "TRP channelopathies," are responsible for a number of hereditary diseases that affect the musculoskeletal, cardiovascular, genitourinary, and nervous systems. This review gives an overview of the functional properties of mammalian TRP channels, describes their roles in acquired and hereditary diseases, and discusses their potential as drug targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bernd Nilius
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| | - Arpad Szallasi
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| |
Collapse
|
50
|
Morales E, Groom A, Lawlor DA, Relton CL. DNA methylation signatures in cord blood associated with maternal gestational weight gain: results from the ALSPAC cohort. BMC Res Notes 2014; 7:278. [PMID: 24886386 PMCID: PMC4108052 DOI: 10.1186/1756-0500-7-278] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 04/10/2014] [Indexed: 12/17/2022] Open
Abstract
Background Epigenetic changes could mediate the association of maternal pre-pregnancy body mass index (BMI) and gestational weight gain (GWG) with adverse offspring outcomes. However, studies in humans are lacking. Here, we examined the association of maternal pre-pregnancy BMI and GWG in different periods of pregnancy with cytosine-guanine (CpG) dinucleotide site methylation differences in newborn cord blood DNA from 88 participants in the Avon Longitudinal Study of Parents and Children (ALSPAC) cohort using the Illumina GoldenGate Panel I. Pyrosequencing was used for validation of the top associated locus and for replication in 170 non-overlapping mother-offspring pairs from the ALSPAC cohort. Results After correction for multiple testing greater GWG in early pregnancy (between 0 to 18 weeks of gestation) was associated with increased DNA methylation levels in four CpG sites at MMP7, KCNK4, TRPM5 and NFKB1 genes (difference in methylation >5% per 400 g/week greater GWG) (q values 0.023 -0.065). Pre-pregnancy BMI and GWG in mid- or late pregnancy were not associated with differential DNA methylation at any CpG site. Pyrosequencing showed that greater GWG in early pregnancy was associated with increased DNA methylation levels at the top associated CpG site at MMP7, although association did not reach statistical significance (p = 0.302). Greater GWG in mid- (p = 0.167) and late-pregnancy (p = 0.037) were also associated with increased DNA methylation levels at the MMP7 CpG site. In addition, newborns of mothers who exceeded the IoM-recommended GWG had higher DNA methylation levels at the MMP7 CpG site than those of mothers with IoM-recommended GWG (p = 0.080). We failed to replicate findings. Conclusions Greater GWG in early pregnancy was associated with increased methylation at CpG sites at MMP7, KCNK4, TRPM5 and NFKB1 genes in offspring cord blood DNA. The specific association of GWG in early pregnancy with the top associated CpG site at MMP7 was not validated using Pyrosequencing and it did not replicate. However, given the potential functional relevancy of the four identified loci, we advocate further exploration of them in larger studies.
Collapse
Affiliation(s)
| | | | | | - Caroline L Relton
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, Tyne and Wear, UK.
| |
Collapse
|