1
|
Chen Y, Jia J, Zhao Q, Zhang Y, Huang B, Wang L, Tian J, Huang C, Li M, Li X. Novel Loss-of-Function Variant in HNF1a Induces β-Cell Dysfunction through Endoplasmic Reticulum Stress. Int J Mol Sci 2022; 23:ijms232113022. [PMID: 36361808 PMCID: PMC9656704 DOI: 10.3390/ijms232113022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 12/02/2022] Open
Abstract
Heterozygous variants in the hepatocyte nuclear factor 1a (HNF1a) cause MODY3 (maturity-onset diabetes of the young, type 3). In this study, we found a case of novel HNF1a p.Gln125* (HNF1a-Q125ter) variant clinically. However, the molecular mechanism linking the new HNF1a variant to impaired islet β-cell function remains unclear. Firstly, a similar HNF1a-Q125ter variant in zebrafish (hnf1a+/−) was generated by CRISPR/Cas9. We further crossed hnf1a+/− with several zebrafish reporter lines to investigate pancreatic β-cell function. Next, we introduced HNF1a-Q125ter and HNF1a shRNA plasmids into the Ins-1 cell line and elucidated the molecular mechanism. hnf1a+/− zebrafish significantly decreased the β-cell number, insulin expression, and secretion. Moreover, β cells in hnf1a+/− dilated ER lumen and increased the levels of ER stress markers. Similar ER-stress phenomena were observed in an HNF1a-Q125ter-transfected Ins-1 cell. Follow-up investigations demonstrated that HNF1a-Q125ter induced ER stress through activating the PERK/eIF2a/ATF4 signaling pathway. Our study found a novel loss-of-function HNF1a-Q125ter variant which induced β-cell dysfunction by activating ER stress via the PERK/eIF2a/ATF4 signaling pathway.
Collapse
Affiliation(s)
- Yinling Chen
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jianxin Jia
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Qing Zhao
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yuxian Zhang
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
| | - Bingkun Huang
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
| | - Likun Wang
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Juanjuan Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Caoxin Huang
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences and School of Life Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (M.L.); (X.L.)
| | - Xuejun Li
- Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
- Fujian Province Key Laboratory of Diabetes Translational Medicine, Xiamen Diabetes Institute, Xiamen 361003, China
- Correspondence: (M.L.); (X.L.)
| |
Collapse
|
2
|
Is Type 2 Diabetes a Primary Mitochondrial Disorder? Cells 2022; 11:cells11101617. [PMID: 35626654 PMCID: PMC9140179 DOI: 10.3390/cells11101617] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/27/2022] [Accepted: 04/20/2022] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is the most common endocrine disturbance in inherited mitochondrial diseases. It is essential to increase awareness of the correct diagnosis and treatment of diabetes in these patients and screen for the condition in family members, as diabetes might appear with distinctive clinical features, complications and at different ages of onset. The severity of mitochondrial-related diabetes is likely to manifest on a large scale of phenotypes depending on the location of the mutation and whether the number of affected mitochondria copies (heteroplasmy) reaches a critical threshold. Regarding diabetes treatment, the first-choice treatment for type 2 diabetes (T2D), metformin, is not recommended because of the risk of lactic acidosis. The preferred treatment for diabetes in patients with mitochondrial disorders is SGLT-2i and mitochondrial GLP-1-related substances. The tight relationship between mitochondrial dysfunction, reduced glucose-stimulated insulin secretion (GSIS), and diabetes development in human patients is acknowledged. However, despite the well-characterized role of mitochondria in GSIS, there is a relative lack of data in humans implicating mitochondrial dysfunction as a primary defect in T2D. Our recent studies have provided data supporting the significant role of the mitochondrial respiratory-chain enzyme, cytochrome c oxidase (COX), in regulating GSIS in a rodent model of T2D, the Cohen diabetic sensitive (CDs) rat. The nutritionally induced diabetic CDs rat demonstrates several features of mitochondrial diseases: markedly reduced COX activity in several tissues, increased reactive oxygen production, decreased ATP generation, and increased lactate dehydrogenase expression in islets. Moreover, our data demonstrate that reduced islet-COX activity precedes the onset of diabetes, suggesting that islet-COX deficiency is the primary defect causing diabetes in this model. This review examines the possibility of including T2D as a primary mitochondrial-related disease. Understanding the critical interdependence between diabetes and mitochondrial dysfunction, centering on the role of COX, may open novel avenues to diagnose and treat diabetes in patients with mitochondrial diseases and mitochondrial dysfunction in diabetic patients.
Collapse
|
3
|
Sanchez Caballero L, Gorgogietas V, Arroyo MN, Igoillo-Esteve M. Molecular mechanisms of β-cell dysfunction and death in monogenic forms of diabetes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:139-256. [PMID: 33832649 DOI: 10.1016/bs.ircmb.2021.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monogenetic forms of diabetes represent 1%-5% of all diabetes cases and are caused by mutations in a single gene. These mutations, that affect genes involved in pancreatic β-cell development, function and survival, or insulin regulation, may be dominant or recessive, inherited or de novo. Most patients with monogenic diabetes are very commonly misdiagnosed as having type 1 or type 2 diabetes. The severity of their symptoms depends on the nature of the mutation, the function of the affected gene and, in some cases, the influence of additional genetic or environmental factors that modulate severity and penetrance. In some patients, diabetes is accompanied by other syndromic features such as deafness, blindness, microcephaly, liver and intestinal defects, among others. The age of diabetes onset may also vary from neonatal until early adulthood manifestations. Since the different mutations result in diverse clinical presentations, patients usually need different treatments that range from just diet and exercise, to the requirement of exogenous insulin or other hypoglycemic drugs, e.g., sulfonylureas or glucagon-like peptide 1 analogs to control their glycemia. As a consequence, awareness and correct diagnosis are crucial for the proper management and treatment of monogenic diabetes patients. In this chapter, we describe mutations causing different monogenic forms of diabetes associated with inadequate pancreas development or impaired β-cell function and survival, and discuss the molecular mechanisms involved in β-cell demise.
Collapse
Affiliation(s)
- Laura Sanchez Caballero
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Vyron Gorgogietas
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Maria Nicol Arroyo
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/.
| |
Collapse
|
4
|
Jesinkey SR, Madiraju AK, Alves TC, Yarborough OH, Cardone RL, Zhao X, Parsaei Y, Nasiri AR, Butrico G, Liu X, Molina AJ, Rountree AM, Neal AS, Wolf DM, Sterpka J, Philbrick WM, Sweet IR, Shirihai OH, Kibbey RG. Mitochondrial GTP Links Nutrient Sensing to β Cell Health, Mitochondrial Morphology, and Insulin Secretion Independent of OxPhos. Cell Rep 2019; 28:759-772.e10. [PMID: 31315053 PMCID: PMC6713209 DOI: 10.1016/j.celrep.2019.06.058] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 02/15/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022] Open
Abstract
Mechanisms coordinating pancreatic β cell metabolism with insulin secretion are essential for glucose homeostasis. One key mechanism of β cell nutrient sensing uses the mitochondrial GTP (mtGTP) cycle. In this cycle, mtGTP synthesized by succinyl-CoA synthetase (SCS) is hydrolyzed via mitochondrial PEPCK (PEPCK-M) to make phosphoenolpyruvate, a high-energy metabolite that integrates TCA cycling and anaplerosis with glucose-stimulated insulin secretion (GSIS). Several strategies, including xenotopic overexpression of yeast mitochondrial GTP/GDP exchanger (GGC1) and human ATP and GTP-specific SCS isoforms, demonstrated the importance of the mtGTP cycle. These studies confirmed that mtGTP triggers and amplifies normal GSIS and rescues defects in GSIS both in vitro and in vivo. Increased mtGTP synthesis enhanced calcium oscillations during GSIS. mtGTP also augmented mitochondrial mass, increased insulin granule number, and membrane proximity without triggering de-differentiation or metabolic fragility. These data highlight the importance of the mtGTP signal in nutrient sensing, insulin secretion, mitochondrial maintenance, and β cell health.
Collapse
Affiliation(s)
- Sean R Jesinkey
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Anila K Madiraju
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA; Departments of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Tiago C Alves
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - OrLando H Yarborough
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Rebecca L Cardone
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA; Departments of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Xiaojian Zhao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Yassmin Parsaei
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ali R Nasiri
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Gina Butrico
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Anthony J Molina
- Division of Geriatrics and Gerontology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Austin M Rountree
- University of Washington Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Adam S Neal
- University of Washington Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Dane M Wolf
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Departments of Medicine, Endocrinology, and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - John Sterpka
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - William M Philbrick
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ian R Sweet
- University of Washington Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Orian H Shirihai
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Departments of Medicine, Endocrinology, and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Richard G Kibbey
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA; Departments of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
5
|
Jungtrakoon P, Shirakawa J, Buranasupkajorn P, Gupta MK, De Jesus DF, Pezzolesi MG, Panya A, Hastings T, Chanprasert C, Mendonca C, Kulkarni RN, Doria A. Loss-of-Function Mutation in Thiamine Transporter 1 in a Family With Autosomal Dominant Diabetes. Diabetes 2019; 68:1084-1093. [PMID: 30833467 PMCID: PMC6477897 DOI: 10.2337/db17-0821] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 02/23/2019] [Indexed: 01/19/2023]
Abstract
Solute Carrier Family 19 Member 2 (SLC19A2) encodes thiamine transporter 1 (THTR1), which facilitates thiamine transport across the cell membrane. SLC19A2 homozygous mutations have been described as a cause of thiamine-responsive megaloblastic anemia (TRMA), an autosomal recessive syndrome characterized by megaloblastic anemia, diabetes, and sensorineural deafness. Here we describe a loss-of-function SLC19A2 mutation (c.A1063C: p.Lys355Gln) in a family with early-onset diabetes and mild TRMA traits transmitted in an autosomal dominant fashion. We show that SLC19A2-deficient β-cells are characterized by impaired thiamine uptake, which is not rescued by overexpression of the p.Lys355Gln mutant protein. We further demonstrate that SLC19A2 deficit causes impaired insulin secretion in conjunction with mitochondrial dysfunction, loss of protection against oxidative stress, and cell cycle arrest. These findings link SLC19A2 mutations to autosomal dominant diabetes and suggest a role of SLC19A2 in β-cell function and survival.
Collapse
Affiliation(s)
- Prapaporn Jungtrakoon
- Department of Medicine, Harvard Medical School, Boston, MA
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, MA
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Jun Shirakawa
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Patinut Buranasupkajorn
- Department of Medicine, Harvard Medical School, Boston, MA
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, MA
- Division of Hospital and Ambulatory Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Manoj K Gupta
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Dario F De Jesus
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Marcus G Pezzolesi
- Division of Nephrology and Hypertension, University of Utah School of Medicine, Salt Lake City, UT
| | - Aussara Panya
- Department of Medicine, Harvard Medical School, Boston, MA
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, MA
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Timothy Hastings
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, MA
| | - Chutima Chanprasert
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Christine Mendonca
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, MA
| | - Rohit N Kulkarni
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Molecular Medicine, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Alessandro Doria
- Department of Medicine, Harvard Medical School, Boston, MA
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, MA
| |
Collapse
|
6
|
Casemayou A, Fournel A, Bagattin A, Schanstra J, Belliere J, Decramer S, Marsal D, Gillet M, Chassaing N, Huart A, Pontoglio M, Knauf C, Bascands JL, Chauveau D, Faguer S. Hepatocyte Nuclear Factor-1 β Controls Mitochondrial Respiration in Renal Tubular Cells. J Am Soc Nephrol 2017; 28:3205-3217. [PMID: 28739648 DOI: 10.1681/asn.2016050508] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/18/2017] [Indexed: 12/19/2022] Open
Abstract
AKI is a frequent condition that involves renal microcirculation impairment, infiltration of inflammatory cells with local production of proinflammatory cytokines, and subsequent epithelial disorders and mitochondrial dysfunction. Peroxisome proliferator-activated receptor γ coactivator 1-α (PPARGC1A), a coactivator of the transcription factor PPAR-γ that controls mitochondrial biogenesis and function, has a pivotal role in the early dysfunction of the proximal tubule and the subsequent renal repair. Here, we evaluated the potential role of hepatocyte nuclear factor-1β (HNF-1β) in regulating PPARGC1A expression in AKI. In mice, endotoxin injection to induce AKI also induced early and transient inflammation and PPARGC1A inhibition, which overlapped with downregulation of the HNF-1β transcriptional network. In vitro, exposure of proximal tubule cells to the inflammatory cytokines IFN-γ and TNF-α led to inhibition of HNF-1β transcriptional activity. Moreover, inhibition of HNF-1β significantly reduced PPARGC1A expression and altered mitochondrial morphology and respiration in proximal tubule cells. Chromatin immunoprecipitation assays and PCR analysis confirmed HNF-1β binding to the Ppargc1a promoter in mouse kidneys. We also demonstrated downregulation of renal PPARGC1A expression in a patient with an HNF1B germinal mutation. Thus, we propose that HNF-1β links extracellular inflammatory signals to mitochondrial dysfunction during AKI partly via PPARGC1A signaling. Our findings further strengthen the view of HNF1B-related nephropathy as a mitochondrial disorder in adulthood.
Collapse
Affiliation(s)
- Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Audren Fournel
- University Toulouse III Paul-Sabatier, Toulouse, France.,Institut National de la Santé et de la Recherche Médicale U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan-BP3028, 31024 Toulouse Cedex 3
| | - Alessia Bagattin
- Laboratoire d'Expression Génique, Développement et Maladies, Département Développement, Reproduction et Cancer, Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| | - Joost Schanstra
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Julie Belliere
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology, Internal Medicine and Hypertension, Center for Rare Renal Diseases, Children' Hospital, University Hospital of Toulouse, Toulouse, France
| | - Dimitri Marsal
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Marion Gillet
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Nicolas Chassaing
- Department of Medical Genetics, Hôpital Purpan, University Hospital of Toulouse, Toulouse, France; and
| | - Antoine Huart
- Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Marco Pontoglio
- Laboratoire d'Expression Génique, Développement et Maladies, Département Développement, Reproduction et Cancer, Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| | - Claude Knauf
- University Toulouse III Paul-Sabatier, Toulouse, France.,Institut National de la Santé et de la Recherche Médicale U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan-BP3028, 31024 Toulouse Cedex 3
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale, U1188, DéTROI (Diabète Athérothrombose Thérapies Réunion Océan Indien), University of La Réunion
| | - Dominique Chauveau
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; .,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
7
|
Abstract
Maturity Onset Diabetes of the Young type 3 (MODY3), linked to mutations in the transcription factor HNF1A, is the most prevalent form of monogenic diabetes mellitus. HNF1alpha-deficiency leads to defective insulin secretion via a molecular mechanism that is still not completely understood. Moreover, in MODY3 patients the severity of insulin secretion can be extremely variable even in the same kindred, indicating that modifier genes may control the onset of the disease. With the use of a mouse model for HNF1alpha-deficiency, we show here that specific genetic backgrounds (C3H and CBA) carry a powerful genetic suppressor of diabetes. A genome scan analysis led to the identification of a major suppressor locus on chromosome 3 (Moda1). Moda1 locus contains 11 genes with non-synonymous SNPs that significantly interacts with other loci on chromosomes 4, 11 and 18. Mechanistically, the absence of HNF1alpha in diabetic-prone (sensitive) strains leads to postnatal defective islets growth that is remarkably restored in resistant strains. Our findings are relevant to human genetics since Moda1 is syntenic with a human locus identified by genome wide association studies of fasting glycemia in patients. Most importantly, our results show that a single genetic locus can completely suppress diabetes in Hnf1a-deficiency.
Collapse
|
8
|
Jonasson ME, Wicklow BA, Sellers EAC, Dolinsky VW, Doucette CA. Exploring the role of the HNF-1αG319S polymorphism in β cell failure and youth-onset type 2 diabetes: Lessons from MODY and Hnf-1α-deficient animal models. Biochem Cell Biol 2015; 93:487-94. [PMID: 26176428 DOI: 10.1139/bcb-2015-0021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The prevalence of youth-onset type 2 diabetes (T2D) is rapidly increasing worldwide, disproportionately affecting Indigenous youth with Oji-Cree heritage from central Canada. Candidate gene screening has uncovered a novel and private polymorphism in the Oji-Cree population in the hepatocyte nuclear factor-1 alpha (HNF-1α) gene, where a highly conserved glycine residue at position 319 is changed to a serine (termed HNF-1αG319S or simply G319S). Oji-Cree youth who carry one or two copies of the "S-allele" present at diagnosis with less obesity, reduced indicators of insulin resistance, and lower plasma insulin levels at diagnosis, suggestive of a primary defect in the insulin-secreting β cells. Few studies on the impact of the HNF-1αG319S variant on β cell function have been performed to date; however, much can be learned from other clinical phenotypes of HNF-1α-deficiency, including HNF-1α mutations that cause maturity-onset diabetes of the young 3 (MODY3). In addition, evaluation of Hnf-1α-deficient murine models reveals that HNF-1α plays a central role in the regulation of insulin secretion by regulating the expression of key genes involved in β cell glucose-sensing, mitochondrial function, and the maintenance of the β cell phenotype in differentiated β cells. The overall goal of this minireview is to explore the impact of HNF-1α-deficiency on the β cell to better inform future research into the mechanisms of β cell dysfunction in Oji-Cree youth with T2D.
Collapse
Affiliation(s)
- Michael E Jonasson
- d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Brandy A Wicklow
- b Department of Pediatrics and Child Health, University of Manitoba, CE-208 Childrens Hospital, 840 Sherbrook Street, Health Sciences Centre, Winnipeg, MB R3A 1S1, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Elizabeth A C Sellers
- b Department of Pediatrics and Child Health, University of Manitoba, CE-208 Childrens Hospital, 840 Sherbrook Street, Health Sciences Centre, Winnipeg, MB R3A 1S1, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Vernon W Dolinsky
- c Department of Pharmacology and Therapeutics, A203 Chown Bldg., 753 McDermot Avenue, University of Manitoba, Winnipeg, MB R3E 0T6, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| | - Christine A Doucette
- a College of Medicine, Faculty of Health Sciences, Department of Physiology & Pathophysiology, 432 Basic Medical Sciences Building, 745 Bannatyne Avenue, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.,d Children's Hospital Research Institute of Manitoba, 715 McDermot Avenue, Winnipeg MB R3E 3P4, Canada
| |
Collapse
|
9
|
Abstract
The oversupply of calories and sedentary lifestyle has resulted in a rapid increase of diabetes prevalence worldwide. During the past two decades, lines of evidence suggest that mitochondrial dysfunction plays a key role in the pathophysiology of diabetes. Mitochondria are vital to most of the eukaryotic cells as they provide energy in the form of adenosine triphosphate by oxidative phosphorylation. In addition, mitochondrial function is an integral part of glucose-stimulated insulin secretion in pancreatic β-cells. In the present article, we will briefly review the major functions of mitochondria in regard to energy metabolism, and discuss the genetic and environmental factors causing mitochondrial dysfunction in diabetes. In addition, the pathophysiological role of mitochondrial dysfunction in insulin resistance and β-cell dysfunction are discussed. We argue that mitochondrial dysfunction could be the central defect causing the abnormal glucose metabolism in the diabetic state. A deeper understanding of the role of mitochondria in diabetes will provide us with novel insights in the pathophysiology of diabetes. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2010.00047.x, 2010).
Collapse
Affiliation(s)
| | - Kyong Soo Park
- Departments of Internal Medicine ; Molecular Medicine and Biopharmaceutical Sciences, Seoul National University College of Medicine
| | - Ki-Up Lee
- Department of Internal Medicine, University of Ulsan College of Medicine
| | - Hong Kyu Lee
- Department of Internal Medicine, Eulji University College of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Groschner LN, Alam MR, Graier WF. Metabolism-secretion coupling and mitochondrial calcium activities in clonal pancreatic β-cells. VITAMINS AND HORMONES 2014; 95:63-86. [PMID: 24559914 DOI: 10.1016/b978-0-12-800174-5.00003-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic β-cells are the only cells capable of lowering blood glucose by secreting insulin. The β-cell continuously adjusts its secretory activity to substrate availability in order to keep blood glucose levels within the physiological range--a process called metabolism-secretion coupling. Glucose is readily taken up by the β-cell and broken down into intermediates that fuel oxidative metabolism inside the mitochondria to generate ATP. The resulting increase in the ATP/ADP ratio causes closure of plasma membrane KATP channels, thereby depolarizing the cell and triggering the opening of voltage-gated Ca²⁺ channels. Consequential oscillations of cytosolic Ca²⁺ not only mediate the exocytosis of insulin granules but are also relayed to other subcellular compartments including the mitochondria, where Ca²⁺ is required to accelerate mitochondrial metabolism in response to nutrient stimulation. The mitochondrial Ca²⁺ uptake machinery plays a fundamental role in this feed-forward mechanism that guarantees sustained insulin secretion and, thus, represents a promising therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Lukas N Groschner
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Muhammad Rizwan Alam
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Wolfgang F Graier
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
11
|
Quan X, Das R, Xu S, Cline GW, Wiederkehr A, Wollheim CB, Park KS. Mitochondrial phosphate transport during nutrient stimulation of INS-1E insulinoma cells. Mol Cell Endocrinol 2013; 381:198-209. [PMID: 23939247 DOI: 10.1016/j.mce.2013.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/18/2013] [Accepted: 08/03/2013] [Indexed: 11/15/2022]
Abstract
Here, we have investigated the role of inorganic phosphate (Pi) transport in mitochondria of rat clonal β-cells. In α-toxin-permeabilized INS-1E cells, succinate and glycerol-3-phosphate increased mitochondrial ATP release which depends on exogenous ADP and Pi. In the presence of substrates, addition of Pi caused mitochondrial matrix acidification and hyperpolarisation which promoted ATP export. Dissipation of the mitochondrial pH gradient or pharmacological inhibition of Pi transport blocked the effects of Pi on electrochemical gradient and ATP export. Knock-down of the phosphate transporter PiC, however, neither prevented Pi-induced mitochondrial activation nor glucose-induced insulin secretion. Using (31)P NMR we observed reduction of Pi pools during nutrient stimulation of INS-1E cells. Interestingly, Pi loss was less pronounced in mitochondria than in the cytosol. We conclude that matrix alkalinisation is necessary to maintain a mitochondrial Pi pool, at levels sufficient to stimulate energy metabolism in insulin-secreting cells beyond its role as a substrate for ATP synthesis.
Collapse
Affiliation(s)
- Xianglan Quan
- Department of Physiology and Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
12
|
Urocortin 2 autocrine/paracrine and pharmacologic effects to activate AMP-activated protein kinase in the heart. Proc Natl Acad Sci U S A 2013; 110:16133-8. [PMID: 24043794 DOI: 10.1073/pnas.1312775110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Urocortin 2 (Ucn2), a peptide of the corticotropin-releasing factor (CRF) family, binds with high affinity to type 2 CRF receptors (CRFR2) on cardiomyocytes and confers protection against ischemia/reperfusion. The mechanisms by which the Ucn2-CRFR2 axis mitigates against ischemia/reperfusion injury remain incompletely delineated. Activation of AMP-activated protein kinase (AMPK) also limits cardiac damage during ischemia/reperfusion. AMPK is classically activated by alterations in cellular energetics; however, hormones, cytokines, and additional autocrine/paracrine factors also modulate its activity. We examined the effects of both the endogenous cardiac Ucn2 autocrine/paracrine pathway and Ucn2 treatment on AMPK regulation. Ucn2 treatment increased AMPK activation and downstream acetyl-CoA carboxylase phosphorylation and glucose uptake in isolated heart muscles. These actions were blocked by the CRFR2 antagonist anti-sauvagine-30 and by a PKCε translocation-inhibitor peptide (εV1-2). Hypoxia-induced AMPK activation was also blunted in heart muscles by preincubation with either anti-sauvagine-30, a neutralizing anti-Ucn2 antibody, or εV1-2. Treatment with Ucn2 in vivo augmented ischemic AMPK activation and reduced myocardial injury and cardiac contractile dysfunction after regional ischemia/reperfusion in mice. Ucn2 also directly activated AMPK in ex vivo-perfused mouse hearts and diminished injury and contractile dysfunction during ischemia/reperfusion. Thus, both Ucn2 treatment and the endogenous cardiac Ucn2 autocrine/paracrine pathway activate AMPK signaling pathway, via a PKCε-dependent mechanism, defining a Ucn2-CRFR2-PKCε-AMPK pathway that mitigates against ischemia/reperfusion injury.
Collapse
|
13
|
Tian L, Kim HS, Kim H, Jin X, Jung HS, Park KS, Cho KW, Park S, Moon WK. Changes in metabolic markers in insulin-producing β-cells during hypoxia-induced cell death as studied by NMR metabolomics. J Proteome Res 2013; 12:3738-45. [PMID: 23795807 DOI: 10.1021/pr400315e] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This study was designed to investigate changes in the metabolites in the intracellular fluid of the pancreatic β-cell line INS-1 to identify potential early and late biomarkers for predicting hypoxia-induced cell death. INS-1 cells were incubated under normoxic conditions (95% air, 5% CO₂) or hypoxic conditions (1% O₂, 5% CO₂, 95% N₂) for 2, 4, 6, 12, or 24 h. The biological changes indicating the process of cell death were analyzed using the MTT assay, flow cytometry, Western blotting, and immunostaining. Changes in the metabolic profiles from cell lysates were identified using ¹H nuclear magnetic resonance (¹H NMR) spectroscopy, and the spectra were analyzed by the multivariate model Orthogonal Projections to Latent Structure-Discriminant Analysis. Cell viability decreased approximately 40% after 12-24 h of hypoxia, coincident with a high level of cleaved caspase-3. A high level of HIF-1α was detected in the 12-24 h hypoxic conditions. The metabolite profiles were altered according to the degree of exposure to hypoxia. A spectral analysis showed significant differences in creatine-containing compounds at the early stage (2-6 h) and taurine-containing compounds at the late stage (12-24 h), with the detection of HIF-1α and cleaved caspase-3 in cells exposed to hypoxia compared to normoxia. Glycerophosphocholine decreased during the early stage hypoxia. The change in taurine- and creatine-containing compounds and choline species could be involved in the β-cell death process as inhibitors or activators of cell death. Our results imply that assessment by ¹H NMR spectroscopy would be a useful tool to predict the cell death process and to identify molecules regulating hypoxia-induced cell death mechanisms.
Collapse
Affiliation(s)
- Lianji Tian
- Department of Biomedical Science, College of Medicine, Medical Research Center, Seoul National University, 101 Daehangno, Jongnogu, Seoul 110-744, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Mueller KR, Balamurugan AN, Cline GW, Pongratz RL, Hooper RL, Weegman BP, Kitzmann JP, Taylor MJ, Graham ML, Schuurman HJ, Papas KK. Differences in glucose-stimulated insulin secretion in vitro of islets from human, nonhuman primate, and porcine origin. Xenotransplantation 2013; 20:75-81. [PMID: 23384163 DOI: 10.1111/xen.12022] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 12/28/2012] [Indexed: 01/15/2023]
Abstract
Porcine islet xenotransplantation is considered a potential cell-based therapy for type 1 diabetes. It is currently being evaluated in diabetic nonhuman primates (NHP) to assess safety and efficacy of the islet product. However, due to a variety of distinct differences between the respective species, including the insulin secretory characteristics of islets, the suitability and predictive value of the preclinical model in the extrapolation to the clinical setting remain a critical issue. Islets isolated from human (n = 3), NHP (n = 2), adult pig (AP, n = 3), and juvenile pig (JP, n = 4) pancreata were perifused with medium at basal glucose (2.5 mm) followed by high glucose (16.7 mm) concentrations. The total glucose-stimulated insulin secretion (GSIS) was calculated from generated insulin secretion profiles. Nonhuman primate islets exhibited GSIS 3-fold higher than AP islets, while AP and JP islets exhibited GSIS 1/3 and 1/30 of human islets, respectively. The insulin content of NHP and AP islets was similar to that of human islets, whereas that of JP islets was 1/5 of human islets. Despite the fact that human, NHP, and AP islets contain similar amounts of insulin, the much higher GSIS for NHP islets than for AP and JP islets suggests the need for increased dosing of islets from JP and AP in pig-to-NHP transplantation. Porcine islet xenotransplantation to humans may require significantly higher dosing given the lower GSIS of AP islets compared to human islets.
Collapse
Affiliation(s)
- Kate R Mueller
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Bogan JS, Xu Y, Hao M. Cholesterol accumulation increases insulin granule size and impairs membrane trafficking. Traffic 2012; 13:1466-80. [PMID: 22889194 DOI: 10.1111/j.1600-0854.2012.01407.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 08/09/2012] [Accepted: 08/13/2012] [Indexed: 11/28/2022]
Abstract
The formation of mature secretory granules is essential for proper storage and regulated release of hormones and neuropeptides. In pancreatic β cells, cholesterol accumulation causes defects in insulin secretion and may participate in the pathogenesis of type 2 diabetes. Using a novel cholesterol analog, we show for the first time that insulin granules are the major sites of intracellular cholesterol accumulation in live β cells. This is distinct from other, non-secretory cell types, in which cholesterol is concentrated in the recycling endosomes and the trans-Golgi network. Excess cholesterol was delivered specifically to insulin granules, which caused granule enlargement and retention of syntaxin 6 and VAMP4 in granule membranes, with concurrent depletion of these proteins from the trans-Golgi network. Clathrin also accumulated in the granules of cholesterol-overloaded cells, consistent with a possible defect in the last stage of granule maturation, during which clathrin-coated vesicles bud from the immature granules. Excess cholesterol also reduced the docking and fusion of insulin granules at the plasma membrane. Together, the data support a model in which cholesterol accumulation in insulin secretory granules impairs the ability of these vesicles to respond to stimuli, and thus reduces insulin secretion.
Collapse
Affiliation(s)
- Jonathan S Bogan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
16
|
Supale S, Li N, Brun T, Maechler P. Mitochondrial dysfunction in pancreatic β cells. Trends Endocrinol Metab 2012; 23:477-87. [PMID: 22766318 DOI: 10.1016/j.tem.2012.06.002] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 06/02/2012] [Accepted: 06/02/2012] [Indexed: 12/17/2022]
Abstract
In pancreatic β cells, mitochondria play a central role in coupling glucose metabolism to insulin exocytosis, thereby ensuring strict control of glucose-stimulated insulin secretion. Defects in mitochondrial function impair this metabolic coupling, and ultimately promote apoptosis and β cell death. Various factors have been identified that may contribute to mitochondrial dysfunction. In this review we address the emerging concept of complex links between these factors. We also discuss the role of the mitochondrial genome and mutations associated with diabetes, the effect of oxidative stress and reactive oxygen species, the sensitivity of mitochondria to lipotoxicity, and the adaptive dynamics of mitochondrial morphology. Better comprehension of the molecular mechanisms contributing to mitochondrial dysfunction will help drive the development of effective therapeutic approaches.
Collapse
Affiliation(s)
- Sachin Supale
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, 1211 Geneva 4, Switzerland
| | | | | | | |
Collapse
|
17
|
Abstract
HNF1A-maturity onset diabetes of the young (HNF1A-MODY) is caused by mutations in Hnf1a gene encoding the transcription factor hepatocyte nuclear factor 1alpha (HNF1A). An increased rate of apoptosis has been associated with the decrease in beta-cell mass that is a hallmark of HNF1A-MODY and other forms of diabetes. In a cellular model of HNF1A-MODY, we have recently shown that signalling through mammalian target of rapamycin (mTOR) is decreased by the overexpression of a dominant-negative mutant of HNF1A (DN-HNF1A). mTOR is a protein kinase which has important roles in cell metabolism and growth, but also in cell survival, where it has been shown to be both protective and detrimental. Here, we show that pharmacological inhibition of mTOR activity with rapamycin protected INS-1 cells against DN-HNF1A-induced apoptosis. Rapamycin also prevented DN-HNF1A-induced activation of AMP-activated protein kinase (AMPK), an intracellular energy sensor which we have previously shown to mediate DN-HNF1A-induced apoptosis. Conversely, activation of mTOR with leucine potentiated DN-HNF1A-induced apoptosis. Gene silencing of raptor (regulatory associated protein of mTOR), a subunit of mTOR complex 1 (mTORC1), also conferred protection on INS-1 cells against DN-HNF1A-induced apoptosis, confirming that mTORC1 mediates the protective effect. The potential relevance of this effect with regards to the clinical use of rapamycin as an immunosuppressant in diabetics post-transplantation is discussed.
Collapse
|
18
|
Suszynski TM, Rizzari MD, Kidder LS, Mueller K, Chapman CS, Kitzmann JP, Pongratz RL, Cline GW, Todd PW, Kennedy DJ, O'Brien TD, Avgoustiniatos ES, Schuurman HJ, Papas KK. Paramagnetic microparticles do not elicit islet cytotoxicity with co-culture or host immune reactivity after implantation. Xenotransplantation 2012; 18:239-44. [PMID: 21848541 DOI: 10.1111/j.1399-3089.2011.00648.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Paramagnetic microparticles (MPs) may be useful in pancreatic islet purification, in particular purification of porcine islets as a potential xenotransplantation product. We assessed whether MPs affect islet function or induce an adverse effect following implantation. METHODS Porcine islets were co-cultured with 0, 500, and 1500 MPs per islet equivalent (IE) for 1 day and with 0 and 1500 MPs/IE for 7 days. Fractional viability was assessed using oxygen consumption rate normalized to DNA content (OCR/DNA) and after 7-day co-culture by perifusion glucose-stimulated insulin secretion (GSIS) and by transplantation under the renal capsule of diabetic nude mice. To assess an inflammatory response or immune reaction, MPs (∼10(7)) were implanted under the renal capsule of C57BL/6 mice. RESULTS No statistically significant differences were measured in OCR/DNA (mean ± SE) following 1-day co-culture with 0, 500, or 1500 MPs/IE (243.3 ± 4.5, 211.3 ± 8.1, or 230.6 ± 11.3 nmol/min·mgDNA, respectively) or following 7-day co-culture with 0 or 1500 MPs/IE (248.5 ± 1.4 or 252.9 ± 4.7 nmol/min·mgDNA, respectively). GSIS was not affected by the presence of MPs; first- and second-phase insulin area-under-the-curve (mean ± SE) reflected no statistically significant differences after 7-day co-culture between 0 and 1500 MPs/IE (8.36 ± 0.29 and 8.45 ± 0.70 pg/ml·min·ngDNA for first-phase; 69.73 ± 2.18 and 65.70 ± 4.34 pg/ml·min·ngDNA for second-phase, respectively). Islets co-cultured with MPs normalized hyperglycemia in diabetic nude mice, suggesting no adverse effects on in vivo islet function. Implantation of MPs did not elicit tissue injury, inflammatory change or immune reactivity. CONCLUSION MPs do not adversely affect islet viability or function during co-culture, and MPs are not immune reactive following implantation.
Collapse
Affiliation(s)
- Thomas M Suszynski
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN 55401, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Cline GW, Pongratz RL, Zhao X, Papas KK. Rates of insulin secretion in INS-1 cells are enhanced by coupling to anaplerosis and Kreb's cycle flux independent of ATP synthesis. Biochem Biophys Res Commun 2011; 415:30-5. [PMID: 22008547 DOI: 10.1016/j.bbrc.2011.09.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 09/30/2011] [Indexed: 01/08/2023]
Abstract
Mechanistic models of glucose stimulated insulin secretion (GSIS) established in minimal media in vitro, may not accurately describe the complexity of coupling metabolism with insulin secretion that occurs in vivo. As a first approximation, we have evaluated metabolic pathways in a typical growth media, DMEM as a surrogate in vivo medium, for comparison to metabolic fluxes observed under the typical experimental conditions using the simple salt-buffer of KRB. Changes in metabolism in response to glucose and amino acids and coupling to insulin secretion were measured in INS-1 832/13 cells. Media effects on mitochondrial function and the coupling efficiency of oxidative phosphorylation were determined by fluorometrically measured oxygen consumption rates (OCRs) combined with (31)P NMR measured rates of ATP synthesis. Substrate preferences and pathways into the TCA cycle, and the synthesis of mitochondrial 2nd messengers by anaplerosis were determined by (13)C NMR isotopomer analysis of the fate of [U-(13)C] glucose metabolism. Despite similar incremental increases in insulin secretion, the changes of OCR in response to increasing glucose from 2.5 to 15mM were blunted in DMEM relative to KRB. Basal and stimulated rates of insulin secretion rates were consistently higher in DMEM, while ATP synthesis rates were identical in both DMEM and KRB, suggesting greater mitochondrial uncoupling in DMEM. The relative rates of anaplerosis, and hence synthesis and export of 2nd messengers from the mitochondria were found to be similar in DMEM to those in KRB. And, the correlation of total PC flux with insulin secretion rates in DMEM was found to be congruous with the correlation in KRB. Together, these results suggest that signaling mechanisms associated with both TCA cycle flux and with anaplerotic flux, but not ATP production, may be responsible for the enhanced rates of insulin secretion in more complex, and physiologically-relevant media.
Collapse
Affiliation(s)
- Gary W Cline
- The Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | |
Collapse
|
20
|
Cline GW. Fuel-Stimulated Insulin Secretion Depends upon Mitochondria Activation and the Integration of Mitochondrial and Cytosolic Substrate Cycles. Diabetes Metab J 2011; 35:458-65. [PMID: 22111036 PMCID: PMC3221020 DOI: 10.4093/dmj.2011.35.5.458] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The pancreatic islet β-cell is uniquely specialized to couple its metabolism and rates of insulin secretion with the levels of circulating nutrient fuels, with the mitochondrial playing a central regulatory role in this process. In the β-cell, mitochondrial activation generates an integrated signal reflecting rates of oxidativephosphorylation, Kreb's cycle flux, and anaplerosis that ultimately determines the rate of insulin exocytosis. Mitochondrial activation can be regulated by proton leak and mediated by UCP2, and by alkalinization to utilize the pH gradient to drive substrate and ion transport. Converging lines of evidence support the hypothesis that substrate cycles driven by rates of Kreb's cycle flux and by anaplerosis play an integral role in coupling responsive changes in mitochondrial metabolism with insulin secretion. The components and mechanisms that account for the integrated signal of ATP production, substrate cycling, the regulation of cellular redox state, and the production of other secondary signaling intermediates are operative in both rodent and human islet β-cells.
Collapse
Affiliation(s)
- Gary W. Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
Kirkpatrick CL, Wiederkehr A, Baquié M, Akhmedov D, Wang H, Gauthier BR, Akerman I, Ishihara H, Ferrer J, Wollheim CB. Hepatic nuclear factor 1alpha (HNF1alpha) dysfunction down-regulates X-box-binding protein 1 (XBP1) and sensitizes beta-cells to endoplasmic reticulum stress. J Biol Chem 2011; 286:32300-12. [PMID: 21784843 DOI: 10.1074/jbc.m111.247866] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Correct endoplasmic reticulum (ER) function is critical for the health of secretory cells, such as the pancreatic β-cell, and ER stress is often a contributory factor to β-cell death in type 2 diabetes. We have used an insulin-secreting cell line with inducible expression of dominant negative (DN) HNF1α, a transcription factor vital for correct β-cell development and function, to show that HNF1α is required for Xbp1 transcription and maintenance of the normal ER stress response. DN HNF1α expression sensitizes the β-cell to ER stress by directly down-regulating Xbp1 transcription, whereas Atf6 is unaffected. Furthermore, DN HNF1α alters calcium homeostasis, resulting in elevated cytoplasmic calcium and increased store-operated calcium entry, whereas mitochondrial calcium uptake is normal. Loss of function of XBP1 is toxic to the β-cell and decreases production of the ER chaperone BiP, even in the absence of ER stress. DN HNF1α-induced sensitivity to cyclopiazonic acid can be partially rescued with the chemical chaperone tauroursodeoxycholate. Rat insulin 2 promoter-DN HNF1α mouse islets express lower levels of BiP mRNA, synthesize less insulin, and are sensitized to ER stress relative to matched control mouse islets, suggesting that this mechanism is also operating in vivo.
Collapse
Affiliation(s)
- Clare L Kirkpatrick
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, Université de Genève, 1 Rue Michel-Servet, 1211 Genève 4, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Jurczak MJ, Lee HY, Birkenfeld AL, Jornayvaz FR, Frederick DW, Pongratz RL, Zhao X, Moeckel GW, Samuel VT, Whaley JM, Shulman GI, Kibbey RG. SGLT2 deletion improves glucose homeostasis and preserves pancreatic beta-cell function. Diabetes 2011; 60:890-8. [PMID: 21357472 PMCID: PMC3046850 DOI: 10.2337/db10-1328] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Inhibition of the Na(+)-glucose cotransporter type 2 (SGLT2) is currently being pursued as an insulin-independent treatment for diabetes; however, the behavioral and metabolic consequences of SGLT2 deletion are unknown. Here, we used a SGLT2 knockout mouse to investigate the effect of increased renal glucose excretion on glucose homeostasis, insulin sensitivity, and pancreatic β-cell function. RESEARCH DESIGN AND METHODS SGLT2 knockout mice were fed regular chow or a high-fat diet (HFD) for 4 weeks, or backcrossed onto the db/db background. The analysis used metabolic cages, glucose tolerance tests, euglycemic and hyperglycemic clamps, as well as isolated islet and perifusion studies. RESULTS SGLT2 deletion resulted in a threefold increase in urine output and a 500-fold increase in glucosuria, as well as compensatory increases in feeding, drinking, and activity. SGLT2 knockout mice were protected from HFD-induced hyperglycemia and glucose intolerance and had reduced plasma insulin concentrations compared with controls. On the db/db background, SGLT2 deletion prevented fasting hyperglycemia, and plasma insulin levels were also dramatically improved. Strikingly, prevention of hyperglycemia by SGLT2 knockout in db/db mice preserved pancreatic β-cell function in vivo, which was associated with a 60% increase in β-cell mass and reduced incidence of β-cell death. CONCLUSIONS Prevention of renal glucose reabsorption by SGLT2 deletion reduced HFD- and obesity-associated hyperglycemia, improved glucose intolerance, and increased glucose-stimulated insulin secretion in vivo. Taken together, these data support SGLT2 inhibition as a viable insulin-independent treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Michael J. Jurczak
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
| | - Hui-Young Lee
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
| | - Andreas L. Birkenfeld
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Francois R. Jornayvaz
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - David W. Frederick
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Rebecca L. Pongratz
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Xiaoxian Zhao
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Gilbert W. Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Varman T. Samuel
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jean M. Whaley
- Metabolic Diseases Biology, Bristol-Myers Squibb Research and Development, Princeton, New Jersey
| | - Gerald I. Shulman
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - Richard G. Kibbey
- Department Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
- Corresponding author: Richard G. Kibbey,
| |
Collapse
|
23
|
Kilbride SM, Farrelly AM, Bonner C, Ward MW, Nyhan KC, Concannon CG, Wollheim CB, Byrne MM, Prehn JHM. AMP-activated protein kinase mediates apoptosis in response to bioenergetic stress through activation of the pro-apoptotic Bcl-2 homology domain-3-only protein BMF. J Biol Chem 2010; 285:36199-206. [PMID: 20841353 PMCID: PMC2975242 DOI: 10.1074/jbc.m110.138107] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 08/26/2010] [Indexed: 11/06/2022] Open
Abstract
Heterozygous loss-of-function mutations in the hepatocyte nuclear factor 1A (HNF1A) gene result in the pathogenesis of maturity-onset diabetes-of-the-young type 3, (HNF1A-MODY). This disorder is characterized by a primary defect in metabolism-secretion coupling and decreased beta cell mass, attributed to excessive beta cell apoptosis. Here, we investigated the link between energy stress and apoptosis activation following HNF1A inactivation. This study employed single cell fluorescent microscopy, flow cytometry, gene expression analysis, and gene silencing to study the effects of overexpression of dominant-negative (DN)-HNF1A expression on cellular bioenergetics and apoptosis in INS-1 cells. Induction of DN-HNF1A expression led to reduced ATP levels and diminished the bioenergetic response to glucose. This was coupled with activation of the bioenergetic stress sensor AMP-activated protein kinase (AMPK), which preceded the onset of apoptosis. Pharmacological activation of AMPK using aminoimidazole carboxamide ribonucleotide (AICAR) was sufficient to induce apoptosis in naive cells. Conversely, inhibition of AMPK with compound C or AMPKα gene silencing protected against DN-HNF1A-induced apoptosis. Interestingly, AMPK mediated the induction of the pro-apoptotic Bcl-2 homology domain-3-only protein Bmf (Bcl-2-modifying factor). Bmf expression was also elevated in islets of DN-HNF1A transgenic mice. Furthermore, knockdown of Bmf expression in INS-1 cells using siRNA was sufficient to protect against DN-HNF1A-induced apoptosis. Our study suggests that overexpression of DN-HNF1A induces bioenergetic stress and activation of AMPK. This in turn mediates the transcriptional activation of the pro-apoptotic Bcl-2-homology protein BMF, coupling prolonged energy stress to apoptosis activation.
Collapse
Affiliation(s)
- Seán M. Kilbride
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | | | - Caroline Bonner
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Manus W. Ward
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Kristine C. Nyhan
- the Mater Misericordiae Hospital, Eccles Street, Dublin 7, Ireland, and
| | - Caoimhín G. Concannon
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Claes B. Wollheim
- the Department of Cell Physiology and Metabolism, University Medical Center, CH-1211 Geneva, Switzerland
| | - Maria M. Byrne
- the Mater Misericordiae Hospital, Eccles Street, Dublin 7, Ireland, and
| | - Jochen H. M. Prehn
- From the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
24
|
Holloway GP, Gurd BJ, Snook LA, Lally J, Bonen A. Compensatory increases in nuclear PGC1alpha protein are primarily associated with subsarcolemmal mitochondrial adaptations in ZDF rats. Diabetes 2010; 59:819-28. [PMID: 20103701 PMCID: PMC2844829 DOI: 10.2337/db09-1519] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE We examined in insulin-resistant muscle if, in contrast to long-standing dogma, mitochondrial fatty acid oxidation is increased and whether this is attributed to an increased nuclear content of peroxisome proliferator-activated receptor (PPAR) gamma coactivator (PGC) 1alpha and the adaptations of specific mitochondrial subpopulations. RESEARCH DESIGN AND METHODS Skeletal muscles from male control and Zucker diabetic fatty (ZDF) rats were used to determine 1) intramuscular lipid distribution, 2) subsarcolemmal and intermyofibrillar mitochondrial morphology, 3) rates of palmitate oxidation in subsarcolemmal and intermyofibrillar mitochondria, and 4) the subcellular localization of PGC1alpha. Electotransfection of PGC1alpha cDNA into lean animals tested the notion that increased nuclear PGC1alpha preferentially targeted subsarcolemmal mitochondria. RESULTS Transmission electron microscope analysis revealed that in ZDF animals the number (+50%), width (+69%), and density (+57%) of subsarcolemmal mitochondria were increased (P < 0.05). In contrast, intermyofibrillar mitochondria remained largely unchanged. Rates of palmitate oxidation were approximately 40% higher (P < 0.05) in ZDF subsarcolemmal and intermyofibrillar mitochondria, potentially as a result of the increased PPAR-targeted proteins, carnitine palmitoyltransferase-I, and fatty acid translocase (FAT)/CD36. PGC1alpha mRNA and total protein were not altered in ZDF animals; however, a greater (approximately 70%; P < 0.05) amount of PGC1alpha was located in nuclei. Overexpression of PGC1alpha only increased subsarcolemmal mitochondrial oxidation rates. CONCLUSIONS In ZDF animals, intramuscular lipids accumulate in the intermyofibrillar region (increased size and number), and this is primarily associated with increased oxidative capacity in subsarcolemmal mitochondria (number, size, density, and oxidation rates). These changes may result from an increased nuclear content of PGC1alpha, as under basal conditions, overexpression of PGC1alpha appears to target subsarcolemmal mitochondria.
Collapse
Affiliation(s)
- Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada.
| | | | | | | | | |
Collapse
|