101
|
Abstract
PURPOSE OF REVIEW We provide a review of monogenic diabetes in young children and adolescents with a focus on recognition, management, and pharmacological treatment. RECENT FINDINGS Monogenic forms of diabetes account for approximately 1-2% of diabetes in children and adolescents, and its incidence has increased in recent years due to greater awareness and wider availability of genetic testing. Monogenic diabetes is due to single gene defects that primarily affect beta cell function with more than 30 different genes reported. Children with antibody-negative, C-peptide-positive diabetes should be evaluated and genetically tested for monogenic diabetes. Accurate genetic diagnosis impacts treatment in the most common types of monogenic diabetes, including the use of sulfonylureas in place of insulin or other glucose-lowering agents or discontinuing pharmacologic treatment altogether. Diagnosis of monogenic diabetes can significantly improve patient care by enabling prediction of the disease course and guiding appropriate management and treatment.
Collapse
Affiliation(s)
- May Sanyoura
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, 5841 S. Maryland Ave., MC 1027, Chicago, IL, 60637, USA
| | - Louis H Philipson
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, 5841 S. Maryland Ave., MC 1027, Chicago, IL, 60637, USA
| | - Rochelle Naylor
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago, 5841 S. Maryland Ave., MC 1027, Chicago, IL, 60637, USA.
| |
Collapse
|
102
|
Peng WF, Bai F, Shao K, Shen LS, Li HH, Huang S. The key genes underlying pathophysiology association between the type 2-diabetic and colorectal cancer. J Cell Physiol 2018; 233:8551-8557. [PMID: 29319171 DOI: 10.1002/jcp.26440] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/05/2018] [Indexed: 01/05/2023]
Abstract
Although diabetes mellitus (DM) is reported as an independent risk factor for colorectal cancer (CRC) in many researches, the underlying pathophysiology is still unclear. We investigated the differentially expressed genes (DEGs) for the diabetes and CRC to reveal the underlying pathophysiological association between the type 2-diabetic (T2D) and CRC. Gene expression profiles for T2D (GSE55650), CRC (GSE8671), and Metformin treated cell lines (GSE67342) were downloaded from GEO database. The DEGs between T2D samples and their control samples were identified with t-test and variance analysis. After cluster analysis and functional enrichment analysis, protein-protein interaction (PPI) network was constructed to find potential genes for diabetes and CRC in Metformin's treatment. Totally, we identified 583 overlapped genes, 169 common DEGs, and 414 independent DEGs between T2D and CRC samples. The common genes contained 89 up-regulated (DEGs1) and 80 down-regulated genes (DEGs3); and independent DEGs contained 270 down-regulated genes (DEGs4) in diabetes and 144 down-regulated genes (DEGs2) in CRC. In enrichment analysis, the Ribosome pathway was significantly enriched by the independent DEGs. The common genes were mainly enriched in some inflammatory related pathways. Two target genes of Metformin were significantly interacted with six hub genes (HADHB, NDUFS3, TAF1, MYC, HNFF4A, and MAX) with significant changes in expression values (P < 0.05, t-test). To summary, it is suggested that the six hub genes might play important roles in the process of Metformin treatment for diabetes and CRC. However, specific pathology remains to be further studied.
Collapse
Affiliation(s)
- Wen-Fang Peng
- Department of Endocrinology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Bai
- Department of Endocrinology and Metabolism, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Kan Shao
- Department of Endocrinology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Sha Shen
- Department of Endocrinology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui-Hua Li
- Department of Endocrinology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Huang
- Department of Endocrinology, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
103
|
Muraresku CC, McCormick EM, Falk MJ. Mitochondrial Disease: Advances in clinical diagnosis, management, therapeutic development, and preventative strategies. CURRENT GENETIC MEDICINE REPORTS 2018; 6:62-72. [PMID: 30393588 PMCID: PMC6208355 DOI: 10.1007/s40142-018-0138-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Primary mitochondrial disease encompasses an impressive range of inherited energy deficiency disorders having highly variable molecular etiologies as well as clinical onset, severity, progression, and response to therapies of multi-system manifestations. Significant progress has been made in primary mitochondrial disease diagnostic approaches, clinical management, therapeutic options, and preventative strategies that are tailored to major mitochondrial disease phenotypes and subclasses. RECENT FINDINGS The extensive phenotypic pleiotropy of individual mitochondrial diseases from an organ-based perspective is reviewed. Improved consensus on standards for mitochondrial disease patient care are being complemented by emerging therapies that target specific molecular subtypes of mitochondrial disease. Reproductive counseling options now include preimplantation genetic diagnosis at the time of in vitro fertilization for familial mutations in nuclear genes and some mtDNA disorders. Mitochondrial replacement technologies have promise for some mtDNA disorders, although practical and societal challenges remain to allow their further research analyses and clinical utilization. SUMMARY A dramatic increase has occurred in recent years in the recognition, understanding, treatment options, and preventative strategies for primary mitochondrial disease.
Collapse
Affiliation(s)
- Colleen C. Muraresku
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elizabeth M. McCormick
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Marni J. Falk
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
104
|
Satake H, Saito A, Sakata T. Elucidation of interfacial pH behaviour at the cell/substrate nanogap for in situ monitoring of cellular respiration. NANOSCALE 2018; 10:10130-10136. [PMID: 29781490 DOI: 10.1039/c8nr02950d] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In situ monitoring of cellular metabolism is useful for elucidating dynamic functions of living cells. In our previous studies, cellular respiration was continuously monitored as a change in pH at the cell/electrode nanoscale interface (i.e., interfacial pH) using an ion-sensitive field-effect transistor (ISFET). However, such interfacial pH behaviour on the nanoscale has not been confirmed using other methods such as fluorescence imaging. In this study, we have clarified the interfacial pH behaviour at a cell/substrate nanogap using a laser scanning confocal fluorescence microscope. The phospholipid fluorescein used as a pH indicator was fixed to the plasma membrane on the external side of a cell by inserting its lipophilic alkyl chain into the membrane, and used to observe the change in interfacial pH. As a result, hydrogen ions generated by cellular respiration were gradually accumulated at the cell/substrate nanogap, resulting in a decrease in pH. Moreover, the interfacial pH between the plasma membrane and the substrate became lower than the pH near the surface of cells not in contact with the substrate. The data obtained in this study support the idea that potentiometric ion sensors such as ISFETs can detect a cellular-metabolism-induced change in pH at a cell/electrode nanogap in real time.
Collapse
Affiliation(s)
- Hiroto Satake
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan 113-8656.
| | | | | |
Collapse
|
105
|
Sakata T, Saito A, Sugimoto H. In situ measurement of autophagy under nutrient starvation based on interfacial pH sensing. Sci Rep 2018; 8:8282. [PMID: 29844482 PMCID: PMC5973944 DOI: 10.1038/s41598-018-26719-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 05/18/2018] [Indexed: 12/31/2022] Open
Abstract
In this study, we report a novel method for the in situ measurement of autophagy under nutrient starvation using a principle of semiconductor technology. A semiconductor-based field-effect transistor (FET) biosensor enables the direct detection of ionic or molecular charges under biological conditions. In particular, cellular respiration accompanied by the generation of carbon dioxide can be continuously and directly monitored as a change in pH at a cell/sensor interface. When autophagy was induced in HeLa cells on a FET biosensor under nutrient starvation, the surface potential increased more significantly for about 15 h than that for nonstarved cells. This positive shift indicates an increase in the number of hydrogen ions produced from the respiration of starved cells because the sensing surface was previously designed to be sensitive to pH variation. Therefore, we have found that cellular respiration is more activated by autophagy under nutrient starvation because the amino acids that decomposed from proteins in autophagic cells would have been rapidly spent in cellular respiration.
Collapse
Affiliation(s)
- Toshiya Sakata
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| | - Akiko Saito
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Haruyo Sugimoto
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
106
|
Anupama N, Sindhu G, Raghu KG. Significance of mitochondria on cardiometabolic syndromes. Fundam Clin Pharmacol 2018; 32:346-356. [DOI: 10.1111/fcp.12359] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/12/2018] [Accepted: 02/12/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Nair Anupama
- Agroprocessing and Technology Division; CSIR -National Institute for Interdisciplinary Science and Technology (NIIST); Industrial estate P.O., Pappanamcode Thiruvananthapuram 695019 Kerala India
| | - Ganapathy Sindhu
- Agroprocessing and Technology Division; CSIR -National Institute for Interdisciplinary Science and Technology (NIIST); Industrial estate P.O., Pappanamcode Thiruvananthapuram 695019 Kerala India
| | - Kozhiparambil Gopalan Raghu
- Agroprocessing and Technology Division; CSIR -National Institute for Interdisciplinary Science and Technology (NIIST); Industrial estate P.O., Pappanamcode Thiruvananthapuram 695019 Kerala India
| |
Collapse
|
107
|
Hirose M, Schilf P, Gupta Y, Zarse K, Künstner A, Fähnrich A, Busch H, Yin J, Wright MN, Ziegler A, Vallier M, Belheouane M, Baines JF, Tautz D, Johann K, Oelkrug R, Mittag J, Lehnert H, Othman A, Jöhren O, Schwaninger M, Prehn C, Adamski J, Shima K, Rupp J, Häsler R, Fuellen G, Köhling R, Ristow M, Ibrahim SM. Low-level mitochondrial heteroplasmy modulates DNA replication, glucose metabolism and lifespan in mice. Sci Rep 2018; 8:5872. [PMID: 29651131 PMCID: PMC5897405 DOI: 10.1038/s41598-018-24290-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/29/2018] [Indexed: 01/07/2023] Open
Abstract
Mutations in mitochondrial DNA (mtDNA) lead to heteroplasmy, i.e., the intracellular coexistence of wild-type and mutant mtDNA strands, which impact a wide spectrum of diseases but also physiological processes, including endurance exercise performance in athletes. However, the phenotypic consequences of limited levels of naturally arising heteroplasmy have not been experimentally studied to date. We hence generated a conplastic mouse strain carrying the mitochondrial genome of an AKR/J mouse strain (B6-mtAKR) in a C57BL/6 J nuclear genomic background, leading to >20% heteroplasmy in the origin of light-strand DNA replication (OriL). These conplastic mice demonstrate a shorter lifespan as well as dysregulation of multiple metabolic pathways, culminating in impaired glucose metabolism, compared to that of wild-type C57BL/6 J mice carrying lower levels of heteroplasmy. Our results indicate that physiologically relevant differences in mtDNA heteroplasmy levels at a single, functionally important site impair the metabolic health and lifespan in mice.
Collapse
Affiliation(s)
- Misa Hirose
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Paul Schilf
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Yask Gupta
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Kim Zarse
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Anke Fähnrich
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Junping Yin
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Marvin N Wright
- Institute of Medical Biometry and Statistics, University of Lübeck, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Leibniz Institute for Prevention Research and Epidemiology, BIPS GmbH, Department Biometry and Data Management, Unit Statistical Methods in Genetics and Live-Course Epidemiology, Bremen, Germany
| | | | - Marie Vallier
- Max Planck Institute for Evolutionary Biology, Evolutionary Genomics, Plön, Germany
| | - Meriem Belheouane
- Max Planck Institute for Evolutionary Biology, Evolutionary Genomics, Plön, Germany
| | - John F Baines
- Max Planck Institute for Evolutionary Biology, Evolutionary Genomics, Plön, Germany
- Institute for Experimental Medicine, Section of Evolutionary Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Diethard Tautz
- Max Planck Institute for Evolutionary Biology, Evolutionary Genomics, Plön, Germany
| | - Kornelia Johann
- Center of Brain Behavior & Metabolism, Molecular Endocrinology, University of Lübeck, Lübeck, Germany
| | - Rebecca Oelkrug
- Center of Brain Behavior & Metabolism, Molecular Endocrinology, University of Lübeck, Lübeck, Germany
| | - Jens Mittag
- Center of Brain Behavior & Metabolism, Molecular Endocrinology, University of Lübeck, Lübeck, Germany
| | - Hendrik Lehnert
- Center of Brain Behavior & Metabolism, Clinical Endocrinology and Metabolism, University of Lübeck, Lübeck, Germany
| | - Alaa Othman
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | - Olaf Jöhren
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Markus Schwaninger
- Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | - Cornelia Prehn
- Helmholtz Center, German Research Center for Environmental Health, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | - Jerzy Adamski
- Helmholtz Center, German Research Center for Environmental Health, Institute of Experimental Genetics, Genome Analysis Center, Neuherberg, Germany
| | - Kensuke Shima
- Department of Infectious Disease and Microbiology, University of Lübeck, Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Disease and Microbiology, University of Lübeck, Lübeck, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, Rostock University, Rostock, Germany
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland.
| | - Saleh M Ibrahim
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
108
|
Yeung RO, Hannah-Shmouni F, Niederhoffer K, Walker MA. Not quite type 1 or type 2, what now? Review of monogenic, mitochondrial, and syndromic diabetes. Rev Endocr Metab Disord 2018; 19:35-52. [PMID: 29777474 DOI: 10.1007/s11154-018-9446-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Diabetes mellitus is a heterogeneous group of conditions defined by resultant chronic hyperglycemia. Given the increasing prevalence of diabetes mellitus and the increasing understanding of genetic etiologies, we present a broad review of rare genetic forms of diabetes that have differing diagnostic and/or treatment implications from type 1 and type 2 diabetes. Advances in understanding the genotype-phenotype associations in these rare forms of diabetes offer clinically available examples of evolving precision medicine where defining the correct genetic etiology can radically alter treatment approaches. In this review, we focus on forms of monogenic diabetes, mitochondrial diabetes, and syndromic diabetes.
Collapse
Affiliation(s)
- Roseanne O Yeung
- Division of Endocrinology and Metabolism, University of Alberta, 9114- Clinical Sciences Building, 11350-83 Avenue, Edmonton, AB, T6G 2G3, Canada.
| | - Fady Hannah-Shmouni
- Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Karen Niederhoffer
- Department of Medical Genetics, University of Alberta, 8-53 Medical Sciences Building, Edmonton, AB, T6G 2H7, Canada
| | - Mark A Walker
- Institute of Cellular Medicine (Diabetes), The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
109
|
Chen H, Sun M, Fan Z, Tong M, Chen G, Li D, Ye J, Yang Y, Zhu Y, Zhu J. Mitochondrial C4375T mutation might be a molecular risk factor in a maternal Chinese hypertensive family under haplotype C. Clin Exp Hypertens 2017; 40:518-523. [PMID: 29200319 DOI: 10.1080/10641963.2017.1403622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Here, we reported a Han Chinese essential hypertensive pedigree based on clinical hereditary and molecular data. To know the molecular basis on this family, mitochondrial genome of one proband from the family was identified through direct sequencing analysis. The age of onset year and affected degree of patients are different in this family. And matrilineal family members carrying C4375T mutation and belong to Eastern Asian halopgroup C. Phylogenetic analysis shows 4375C is highly conservative in 17 species. It is suggested that these mutations might participate in the development of hypertension in this family. And halopgroup C might play a modifying role on the phenotype in this Chinese hypertensive family.
Collapse
Affiliation(s)
- Hong Chen
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Min Sun
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Zhen Fan
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Maoqing Tong
- c Department of Cardiology , Ningbo First Hospital, Ningbo, China.,d Key Laboratory of Molecular Medicine , Ningbo First Hospital , Ningbo , Zhejiang P.R. China
| | - Guodong Chen
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Danhui Li
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Jihui Ye
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Yumin Yang
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Yongding Zhu
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| | - Jianhua Zhu
- a Intensive Care Unit , Ningbo First Hospital, Ningbo, China.,b Intensive Care Unit , Ningbo Hospital of Zhejiang University, Ningbo, China
| |
Collapse
|
110
|
Prudente S, Ludovico O, Trischitta V. Familial diabetes of adulthood: A bin of ignorance that needs to be addressed. Nutr Metab Cardiovasc Dis 2017; 27:1053-1059. [PMID: 29174219 DOI: 10.1016/j.numecd.2017.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/29/2017] [Accepted: 10/15/2017] [Indexed: 01/15/2023]
Abstract
AIMS The aim of this article was to share with a wide readership some data and related reasoning about a multigenerational form of diabetes mellitus of adulthood. DATA SYNTHESIS We have recently described a familial form of diabetes mellitus, which in the routine clinical setting of adult individuals is simplistically diagnosed as type 2 diabetes. Such misdiagnosis involves as much as 3% of adult unrelated diabetic patients with no evidence of autoimmune disease. More recent data, obtained by means of a next-generation sequencing, indicate that approximately 25% of such patients carry mutations in the genes involved in monogenic diabetes, thus leaving unraveled the molecular causes of the remaining 75% individuals. CONCLUSIONS Our proposal is to define the latter patients as being affected by familial diabetes of adulthood (FDA), a clear admission of ignorance and a limbo where adult patients with multigenerational diabetes with no genetic definition of their hyperglycemia have to wait for better times.
Collapse
Affiliation(s)
- S Prudente
- Research Unit of Metabolic and Cardiovascular Diseases, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | - O Ludovico
- Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - V Trischitta
- Research Unit of Metabolic and Cardiovascular Diseases, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy; Department of Experimental Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
111
|
Høgild Langdahl J, Frederiksen AL, Andersen PH, Yderstraede KB, Frost M. Reply to: Reduced Bone Mineral Density in m.3243A>G Carriers May Be Multifactorial. J Bone Miner Res 2017; 32:2317-2318. [PMID: 28850729 DOI: 10.1002/jbmr.3280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jakob Høgild Langdahl
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
| | - Anja Lisbeth Frederiksen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Per Heden Andersen
- Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
| | | | - Morten Frost
- Department of Endocrinology, Odense University Hospital, ODense, Denmark
| |
Collapse
|
112
|
Tabebi M, Khabou B, Boukadi H, Ben Hamad M, Ben Rhouma B, Tounsi S, Maalej A, Kamoun H, Keskes-Ammar L, Abid M, Mnif M, Fakhfakh F. Association study of apoptosis gene polymorphisms in mitochondrial diabetes: A potential role in the pathogenicity of MD. Gene 2017; 639:18-26. [PMID: 28987347 DOI: 10.1016/j.gene.2017.09.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/07/2017] [Accepted: 09/27/2017] [Indexed: 12/27/2022]
Abstract
Mitochondrial diabetes (MD) is a heterogeneous disorder characterized by a chronic hyperglycemia and is maternally transmitted. Syndromic MD is a subgroup of MD including diabetic microangiopathy and macroangiopathy, in addition to extrapancreatic disorder. MD is caused by genetic mutations and deletions affecting mitochondrial DNA. This mitochondrial damage initiates apoptosis. In this study, we hypothesized that functional polymorphisms in genes involved in apoptotic pathway could be associated with the development of apoptosis in MD disease and increased its risk. Detection of apoptosis was confirmed on muscle biopsies taken from MD patients using the TUNEL method and the Cytochrome c protein expression level. We genotyped then 11 published SNPs from intrinsic and extrinsic apoptotic pathway and assessed the signification of these polymorphisms in 43 MD patients and 100 healthy controls. We found 10 selected polymorphisms (p53 (rs1042522 and rs17878362), BCL2 (rs2279115), BAX (rs1805419), BAK1 (rs210132 and rs2227925), CASP3 (rs1405937), CASP7 (rs2227310), CASP8 (rs1045485) and CASP10 (rs13006529)) with a potential apoptosis effect in MD patients compared to control population. Specifically, SNPs involved in the intrinsic pathway (p53, BCL2, BAK1 and CASP3) presented the highest risk of apoptosis. Our result proved that apoptosis initiated by mtDNA mutations, can be emphasized by a functional apoptotic polymorphisms associated with high expression of cytochrome c protein and more myofibers with apoptosis in syndromic MD subgroup compared with non-syndromic MD subgroup.
Collapse
Affiliation(s)
- Mouna Tabebi
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia.
| | - Bodour Khabou
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Hanen Boukadi
- Laboratory of Biopesticides, Centre of Biotechnology of Sfax, University of Sfax, Tunisia
| | - Mariam Ben Hamad
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Bochra Ben Rhouma
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Slim Tounsi
- Laboratory of Biopesticides, Centre of Biotechnology of Sfax, University of Sfax, Tunisia
| | - Abdellatif Maalej
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Hassen Kamoun
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Leila Keskes-Ammar
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Mohamed Abid
- Department of Endocrinology, University Hospital Hedi Chaker, Sfax, Tunisia
| | - Mouna Mnif
- Department of Endocrinology, University Hospital Hedi Chaker, Sfax, Tunisia
| | - Faiza Fakhfakh
- Department of Life Sciences, Faculty of Science of Sfax, University of Sfax, Tunisia
| |
Collapse
|
113
|
Langdahl JH, Frederiksen AL, Hansen SJ, Andersen PH, Yderstraede KB, Dunø M, Vissing J, Frost M. Mitochondrial Point Mutation m.3243A>G Associates With Lower Bone Mineral Density, Thinner Cortices, and Reduced Bone Strength: A Case-Control Study. J Bone Miner Res 2017; 32:2041-2048. [PMID: 28603900 DOI: 10.1002/jbmr.3193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/19/2017] [Accepted: 05/31/2017] [Indexed: 12/17/2022]
Abstract
Mitochondrial dysfunction is associated with several clinical manifestations including diabetes mellitus (DM), neurological disorders, renal and hepatic diseases, and myopathy. Although mitochondrial dysfunction is associated with increased bone resorption and decreased bone formation in mouse models, effects of alterations in mitochondrial function on bone remodeling and mass have not been investigated in humans. We recruited 45 carriers (29 females, 16 males) with the m.3243A>G mutation and healthy controls matched for gender, age, height, and menopausal status. DXA and HRpQCT scans were performed, and bone turnover markers (BTMs) P1NP and CTX were measured. Cases and controls were well matched except for body weight, which was lower in cases (63.6 ± 18.1 kg versus 74.6 ± 14.8 kg, p < 0.01), and manifest DM was present in 25 of 45 cases (none in controls). Bone scans showed lower BMD at the lumbar spine, total hip, and femoral neck in cases. Mean lumbar spine, total hip, and femoral neck T-scores were -1.5, -1.3, and -1.6 in cases, respectively, and -0.8, -0.3, and -0.7 in controls (all p < 0.05). The m.3243A>G mutation was associated with lower BMD, cortical but not trabecular density, cortical thickness, and estimated bone strength. Furthermore, BTMs were lower in the m.3243A>G group before but not after adjustment for DM. The mitochondrial point mutation m.3243A>G was associated with decreased bone mass and strength. Although the coexistence of DM may have influenced bone turnover, the bone phenotype observed in m.3243A>G cases appeared to mirror age-related deterioration in bone, suggesting that mitochondrial dysfunction may cause a premature aging of bone. © 2017 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Jakob Høgild Langdahl
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
| | - Anja Lisbeth Frederiksen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Stinus Jørn Hansen
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Per Heden Andersen
- Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
| | | | - Morten Dunø
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Morten Frost
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
114
|
Permana Maksum I, Saputra SR, Indrayati N, Yusuf M, Subroto T. Bioinformatics Study of m.9053G>A Mutation at the ATP6 Gene in Relation to Type 2 Diabetes Mellitus and Cataract Diseases. Bioinform Biol Insights 2017; 11:1177932217728515. [PMID: 28932107 PMCID: PMC5598796 DOI: 10.1177/1177932217728515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/02/2017] [Indexed: 12/11/2022] Open
Abstract
The mitochondrial disease often associated with various illnesses in relation to the activity of cells metabolites and the synthesis of adenosine triphosphate (ATP), including alteration in the mitochondrial DNA. The mutation of m.9053G>A at the ATP6 gene was found in patients with type 2 diabetes mellitus (DM type 2) and cataract. Therefore, this mutation is predicted to be clinical features of the 2 diseases. ATP6 gene encodes protein subunit of ATPase6, a part of ATP synthase, which is important in the electron transfer and proton translocation in intracellular respiration system. This study aims to investigate the mutation effect of m.9053G>A at the ATP6 gene (S167N) to the structure and function of ATPase6 using bioinformatics method. The structure of ATPase6 was constructed using homology modeling method. The crystal structure of bovine’s ATP synthase (Protein Data Bank ID 5FIL) was used as a template because of high sequence similarity (77%) and coverage (96%) of the input sequence. The effect of mutation was investigated at the proton translocation channel of ATPase6. It is predicted that the channel was disrupted due to changes in electrostatic potential from serine to asparagine. Furthermore, molecular docking suggests that water binding on the proton translocation channel in the S167N mutant was different from the wild type. The result of this study is hoped to be useful in the development of a new genetic marker for DM type 2 and cataract.
Collapse
Affiliation(s)
- Iman Permana Maksum
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | - Sandy Risfi Saputra
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | - Nenden Indrayati
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhammad Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia.,Research Centre of Molecular Biotechnology and Bioinformatics, Universitas Padjadjaran, Bandung, Indonesia
| | - Toto Subroto
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Bandung, Indonesia.,Research Centre of Molecular Biotechnology and Bioinformatics, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
115
|
Meng F, Liu Y, Niu J, Lin W. A novel fluorescent probe with a large Stokes shift for real-time imaging mitochondria in different living cell lines. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.07.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
116
|
Zhu J, Yang P, Liu X, Yan L, Rampersad S, Li F, Li H, Sheng C, Cheng X, Zhang M, Qu S. The clinical characteristics of patients with mitochondrial tRNA Leu(UUR)m.3243A > G mutation: Compared with type 1 diabetes and early onset type 2 diabetes. J Diabetes Complications 2017; 31:1354-1359. [PMID: 28599824 DOI: 10.1016/j.jdiacomp.2017.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 03/09/2017] [Accepted: 04/03/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE This study presents nine patients with mitochondrial tRNA Leu (UUR) m.3243A>G mutation and compares the clinical characteristics and diabetes complications with type 1 diabetes (T1DM) or early onset type 2 diabetes (T2DM). METHODS The study covers 9 patients with MIDD, 33 patients with T1DM and 86 patients (age of onset ≤35years) with early onset T2DM, matched for sex, age at onset of diabetes, duration of diabetes. All patients with MIDD were confirmed as carrying the m.3243A>G mitochondrial DNA mutation. Serum HbA1c, beta-cell function, retinal and renal complications of diabetes, bone metabolic markers, lumbar spine and femoral neck BMD bone mineral density were compared to characterize the clinical features of all patients. RESULTS Nine patients were from five unrelated families, and the mean (SD) onset age of those patients was 31.2±7.2year. Two patients required insulin at presentation, and six patients progressed to insulin requirement after a mean of 7.2years. β-Cell function in the MIDD group was intermediate between T1DM and early-onset T2DM. In MIDD, four patients were diagnosed as diabetic retinopathy (4/9) and five patients (5/9) had macroalbuminuria. The number of patients with diabetic retinopathy and macroalbuminuria in the MIDD group was comparable to T1DM or early-onset T2DM. The rate of osteoporosis (BMD T-score<-2.5 SD) in the patient with MIDD was higher than the T1DM or early-onset T2DM group. CONCLUSION Our study indicates that of the nine subjects with MIDD, three patients (1-II-1, 1-II-3, 1-II-4) who came from the same family had a history of acute pancreatitis. Compared with T1DM or early-onset T2DM matched for sex, age, duration of diabetes, MIDD patients had the highest rate of osteoporosis.
Collapse
MESH Headings
- Adult
- Age of Onset
- Biomarkers/blood
- Biomarkers/urine
- Bone Density
- China/epidemiology
- Deafness/complications
- Deafness/genetics
- Deafness/metabolism
- Deafness/physiopathology
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 1/urine
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/urine
- Diabetic Nephropathies/epidemiology
- Diabetic Retinopathy/epidemiology
- Female
- Glycated Hemoglobin/analysis
- Humans
- Male
- Mitochondrial Diseases/complications
- Mitochondrial Diseases/genetics
- Mitochondrial Diseases/metabolism
- Mitochondrial Diseases/physiopathology
- Osteoporosis/complications
- Osteoporosis/epidemiology
- Pancreatitis/complications
- Pancreatitis/epidemiology
- Point Mutation
- Prevalence
- RNA, Transfer, Leu
- Young Adult
Collapse
Affiliation(s)
- Jie Zhu
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Peng Yang
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Xiang Liu
- Department of Urology, Putuo District People's Hospital, Shanghai 200060, China
| | - Li Yan
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Sharvan Rampersad
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Feng Li
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Hong Li
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Chunjun Sheng
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Xiaoyun Cheng
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Manna Zhang
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China.
| | - Shen Qu
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| |
Collapse
|
117
|
Silvander JSG, Kvarnström SM, Kumari-Ilieva A, Shrestha A, Alam CM, Toivola DM. Keratins regulate β-cell mitochondrial morphology, motility, and homeostasis. FASEB J 2017; 31:4578-4587. [PMID: 28666985 DOI: 10.1096/fj.201700095r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/19/2017] [Indexed: 12/19/2022]
Abstract
Loss of the epithelial intermediate filament protein keratin 8 (K8) in murine β cells leads to irregular insulin vesicles and decreased insulin levels. Because mitochondria are central in glucose-stimulated insulin secretion, the relationship between keratins and β-cell mitochondrial function and morphology was investigated. β cells in murine K8-knockout (K8-/-) islets of Langerhans have increased numbers of mitochondria, which are rounder and have diffuse cristae, as seen by electron microscopy. The mitochondrial network in primary cultured K8-/- β cells is more fragmented compared with K8+/+ mitochondria, correlating with decreased levels of mitofusin 2 and the mitofusin 2- and keratin-binding protein trichoplein. K8-/- β-cell mitochondria have decreased levels of total and mitochondrial cytochrome c, which correlates with a reduction in electron transport complexes I and IV. This provokes loss of mitochondrial membrane potential and reduction of ATP and insulin amount, as seen in K8-/- β cells. Mitochondria in K8 wild-type β cells and MIN6 insulinoma cells overexpressing K8 and 18 are more stationary compared with mitochondria in keratin-deficient cells. In conclusion, keratins, likely through trichoplein-mitofusin interactions, regulate both structural and dynamic functions of β-cell mitochondria, which could have implications for downstream insulin secretion.-Silvander, J. S. G., Kvarnström, S. M., Kumari-Ilieva, A., Shrestha, A., Alam, C. M., Toivola, D. M. Keratins regulate β-cell mitochondrial morphology, motility, and homeostasis.
Collapse
Affiliation(s)
- Jonas S G Silvander
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Sofie M Kvarnström
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Angeli Kumari-Ilieva
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Anup Shrestha
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Catharina M Alam
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Diana M Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| |
Collapse
|
118
|
Glycogénose hépatique : une complication rare du diabète déséquilibré (à propos d’un cas). MEDECINE INTENSIVE REANIMATION 2017. [DOI: 10.1007/s13546-017-1280-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
119
|
Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders - A step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1066-1077. [PMID: 27836629 PMCID: PMC5423868 DOI: 10.1016/j.bbadis.2016.11.010] [Citation(s) in RCA: 1007] [Impact Index Per Article: 125.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/02/2016] [Accepted: 11/03/2016] [Indexed: 01/06/2023]
Abstract
Mitochondria are the powerhouses of the cell and are involved in essential functions of the cell, including ATP production, intracellular Ca2+ regulation, reactive oxygen species production & scavenging, regulation of apoptotic cell death and activation of the caspase family of proteases. Mitochondrial dysfunction and oxidative stress are largely involved in aging, cancer, age-related neurodegenerative and metabolic syndrome. In the last decade, tremendous progress has been made in understanding mitochondrial structure, function and their physiology in metabolic syndromes such as diabetes, obesity, stroke and hypertension, and heart disease. Further, progress has also been made in developing therapeutic strategies, including lifestyle interventions (healthy diet and regular exercise), pharmacological strategies and mitochondria-targeted approaches. These strategies were mainly focused to reduce mitochondrial dysfunction and oxidative stress and to maintain mitochondrial quality in metabolic syndromes. The purpose of our article is to highlight the recent progress on the mitochondrial role in metabolic syndromes and also summarize the progress of mitochondria-targeted molecules as therapeutic targets to treat metabolic syndromes. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Department of Biotechnology and Bioinformatics, Sri Guru Gobind Singh College, Sector-26, Chandigarh 160019, India; Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States.
| | - Gurjit Kaur Bhatti
- UGC Centre of Excellence in Nano applications, Panjab University, UIPS building, Chandigarh 160014, India
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neuroscience & Pharmacology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neurology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, MS 7495, Lubbock, TX 79413, United States
| |
Collapse
|
120
|
El-Hattab AW, Craigen WJ, Scaglia F. Mitochondrial DNA maintenance defects. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1539-1555. [PMID: 28215579 DOI: 10.1016/j.bbadis.2017.02.017] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/31/2017] [Accepted: 02/14/2017] [Indexed: 01/12/2023]
Abstract
The maintenance of mitochondrial DNA (mtDNA) depends on a number of nuclear gene-encoded proteins including a battery of enzymes forming the replisome needed to synthesize mtDNA. These enzymes need to be in balanced quantities to function properly that is in part achieved by exchanging intramitochondrial contents through mitochondrial fusion. In addition, mtDNA synthesis requires a balanced supply of nucleotides that is achieved by nucleotide recycling inside the mitochondria and import from the cytosol. Mitochondrial DNA maintenance defects (MDMDs) are a group of diseases caused by pathogenic variants in the nuclear genes involved in mtDNA maintenance resulting in impaired mtDNA synthesis leading to quantitative (mtDNA depletion) and qualitative (multiple mtDNA deletions) defects in mtDNA. Defective mtDNA leads to organ dysfunction due to insufficient mtDNA-encoded protein synthesis, resulting in an inadequate energy production to meet the needs of affected organs. MDMDs are inherited as autosomal recessive or dominant traits, and are associated with a broad phenotypic spectrum ranging from mild adult-onset ophthalmoplegia to severe infantile fatal hepatic failure. To date, pathogenic variants in 20 nuclear genes known to be crucial for mtDNA maintenance have been linked to MDMDs, including genes encoding enzymes of mtDNA replication machinery (POLG, POLG2, TWNK, TFAM, RNASEH1, MGME1, and DNA2), genes encoding proteins that function in maintaining a balanced mitochondrial nucleotide pool (TK2, DGUOK, SUCLG1, SUCLA2, ABAT, RRM2B, TYMP, SLC25A4, AGK, and MPV17), and genes encoding proteins involved in mitochondrial fusion (OPA1, MFN2, and FBXL4).
Collapse
Affiliation(s)
- Ayman W El-Hattab
- Division of Clinical Genetics and Metabolic Disorders, Pediatrics Department, Tawam Hospital, Al-Ain, United Arab Emirates
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
121
|
Bhatti JS, Kumar S, Vijayan M, Bhatti GK, Reddy PH. Therapeutic Strategies for Mitochondrial Dysfunction and Oxidative Stress in Age-Related Metabolic Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 146:13-46. [PMID: 28253984 DOI: 10.1016/bs.pmbts.2016.12.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mitochondria are complex, intercellular organelles present in the cells and are involved in multiple roles including ATP formation, free radicals generation and scavenging, calcium homeostasis, cellular differentiation, and cell death. Many studies depicted the involvement of mitochondrial dysfunction and oxidative damage in aging and pathogenesis of age-related metabolic disorders and neurodegenerative diseases. Remarkable advancements have been made in understanding the structure, function, and physiology of mitochondria in metabolic disorders such as diabetes, obesity, cardiovascular diseases, and stroke. Further, much progress has been done in the improvement of therapeutic strategies, including lifestyle interventions, pharmacological, and mitochondria-targeted therapeutic approaches. These strategies were mainly focused to reduce the mitochondrial dysfunction caused by oxidative stress and to retain the mitochondrial health in various diseases. In this chapter, we have highlighted the involvement of mitochondrial dysfunction in the pathophysiology of various disorders and recent progress in the development of mitochondria-targeted molecules as therapeutic measures for metabolic disorders.
Collapse
Affiliation(s)
- J S Bhatti
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India.
| | - S Kumar
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - M Vijayan
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - G K Bhatti
- UGC Centre of Excellence in Nano Applications, Panjab University, Chandigarh, India
| | - P H Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
122
|
Abstract
Mitochondria are critical organelles for endocrine health; steroid hormone biosynthesis occurs in these organelles and they provide energy in the form of ATP for hormone production and trafficking. Mitochondrial diseases are multisystem disorders that feature defective oxidative phosphorylation, and are characterized by enormous clinical, biochemical and genetic heterogeneity. To date, mitochondrial diseases have been found to result from >250 monogenic defects encoded across two genomes: the nuclear genome and the ancient circular mitochondrial genome located within mitochondria themselves. Endocrine dysfunction is often observed in genetic mitochondrial diseases and reflects decreased intracellular production or extracellular secretion of hormones. Diabetes mellitus is the most frequently described endocrine disturbance in patients with inherited mitochondrial diseases, but other endocrine manifestations in these patients can include growth hormone deficiency, hypogonadism, adrenal dysfunction, hypoparathyroidism and thyroid disease. Although mitochondrial endocrine dysfunction frequently occurs in the context of multisystem disease, some mitochondrial disorders are characterized by isolated endocrine involvement. Furthermore, additional monogenic mitochondrial endocrine diseases are anticipated to be revealed by the application of genome-wide next-generation sequencing approaches in the future. Understanding the mitochondrial basis of endocrine disturbance is key to developing innovative therapies for patients with mitochondrial diseases.
Collapse
Affiliation(s)
- Jasmine Chow
- Department of Paediatrics, Queen Elizabeth Hospital, 30 Gascoigne Road, Kowloon, Hong Kong, China
| | - Joyeeta Rahman
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - John C Achermann
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul T Dattani
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- Endocrinology Unit, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
| | - Shamima Rahman
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- Metabolic Unit, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
| |
Collapse
|
123
|
Tabebi M, Charfi N, Kallabi F, Alila-Fersi O, Ben Mahmoud A, Tlili A, Keskes-Ammar L, Kamoun H, Abid M, Mnif M, Fakhfakh F. Whole mitochondrial genome screening of a family with maternally inherited diabetes and deafness (MIDD) associated with retinopathy: A putative haplotype associated to MIDD and a novel MT-CO2 m.8241T>G mutation. J Diabetes Complications 2017; 31:253-259. [PMID: 27422531 DOI: 10.1016/j.jdiacomp.2016.06.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 06/12/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
Abstract
Mitochondrial diseases are a clinically heterogeneous group of disorders that arise as a result of dysfunction of the mitochondrial respiratory chain. They can be caused by mutations in both nuclear and mitochondrial DNA. In fact, mitochondrial DNA (mtDNA) defects are known to be associated with a large spectrum of human diseases and patients might present wide range of clinical features with various combinations. Our study reported a Tunisian family with clinical features of maternally inherited diabetes and deafness (MIDD). Accordingly, we performed a whole mitochondrial genome mutational analysis, results revealed a haplotype composed by "A750G, A1438G, G8860A, T12705, T14766C and T16519C", in homoplasmic state, in the mother and transmitted to her daughter and her son. The patient with MIDD2 and retinopathy presented, in addition to this haplotype associated to the MIDD, two de novo variations including a novel one m.8241T>G (p. F219C) in MT-CO2 gene and a known one m.13276G>A (p. M314V) in MT-ND5 gene. The coexistence of these two mutations could explain the retinopathy observed in this patient.
Collapse
Affiliation(s)
- Mouna Tabebi
- Human Molecular Genetics Laboratory, Faculty of Medecine of Sfax, University of Sfax, Tunisia.
| | - Nadia Charfi
- Service of endocrinology, C.H.U. Habib Bourguiba of Sfax, Tunisia
| | - Fakhri Kallabi
- Human Molecular Genetics Laboratory, Faculty of Medecine of Sfax, University of Sfax, Tunisia
| | - Olfa Alila-Fersi
- Human Molecular Genetics Laboratory, Faculty of Medecine of Sfax, University of Sfax, Tunisia
| | - Afif Ben Mahmoud
- Human Molecular Genetics Laboratory, Faculty of Medecine of Sfax, University of Sfax, Tunisia
| | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, UAE
| | - Leila Keskes-Ammar
- Human Molecular Genetics Laboratory, Faculty of Medecine of Sfax, University of Sfax, Tunisia
| | - Hassen Kamoun
- Human Molecular Genetics Laboratory, Faculty of Medecine of Sfax, University of Sfax, Tunisia
| | - Mohamed Abid
- Service of endocrinology, C.H.U. Habib Bourguiba of Sfax, Tunisia
| | - Mouna Mnif
- Service of endocrinology, C.H.U. Habib Bourguiba of Sfax, Tunisia
| | - Faiza Fakhfakh
- Human Molecular Genetics Laboratory, Faculty of Medecine of Sfax, University of Sfax, Tunisia; Department of life Sciences, Faculty of Science of Sfax, University of Sfax, Tunisia.
| |
Collapse
|
124
|
Charoute H, Kefi R, Bounaceur S, Benrahma H, Reguig A, Kandil M, Rouba H, Bakhchane A, Abdelhak S, Barakat A. Novel variants of mitochondrial DNA associated with Type 2 diabetes mellitus in Moroccan population. Mitochondrial DNA A DNA Mapp Seq Anal 2016; 29:9-13. [PMID: 27728995 DOI: 10.1080/24701394.2016.1233530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In this study, we investigated the association of mtDNA variants and haplogroups with Type 2 diabetes (T2D) in Moroccan patients. The Hypervariable Segments 1 of the mtDNA was sequenced in 108 diabetic patients and 97 controls. Association analyses were performed using Fisher's exact test and multivariate logistic regression. The prevalence of five mtDNA variants (C16187T, C16270T, T16172C, A16293G, and C16320T) was significantly higher in cases than in controls. Among these variants, only C16270T (p = .02) and C16320T (p = .03) remains significant after adjusting by age and gender. We showed that C16270T and C16320T variants were strongly associated with increased risk of T2D in Moroccan patients.
Collapse
Affiliation(s)
- Hicham Charoute
- a Institut Pasteur, Human Molecular Genetic Laboratory , Casablanca , Morocco
| | - Rym Kefi
- b Biomedical Genomics and Oncogenetics Laboratory (LR 11 IPT 05) , Institut Pasteur de Tunis, Université de Tunis El Manar , Tunis , Tunisia
| | - Safaa Bounaceur
- a Institut Pasteur, Human Molecular Genetic Laboratory , Casablanca , Morocco
| | - Houda Benrahma
- a Institut Pasteur, Human Molecular Genetic Laboratory , Casablanca , Morocco
| | - Ahmed Reguig
- a Institut Pasteur, Human Molecular Genetic Laboratory , Casablanca , Morocco
| | - Mostafa Kandil
- c Equipe d'Anthropogénétique et Biotechnologies, Faculté des Sciences , Université Chouaïb Doukkali , El Jadida , Morocco
| | - Hassan Rouba
- a Institut Pasteur, Human Molecular Genetic Laboratory , Casablanca , Morocco
| | - Amina Bakhchane
- a Institut Pasteur, Human Molecular Genetic Laboratory , Casablanca , Morocco
| | - Sonia Abdelhak
- b Biomedical Genomics and Oncogenetics Laboratory (LR 11 IPT 05) , Institut Pasteur de Tunis, Université de Tunis El Manar , Tunis , Tunisia
| | - Abdelhamid Barakat
- a Institut Pasteur, Human Molecular Genetic Laboratory , Casablanca , Morocco
| |
Collapse
|
125
|
Chuah JA, Matsugami A, Hayashi F, Numata K. Self-Assembled Peptide-Based System for Mitochondrial-Targeted Gene Delivery: Functional and Structural Insights. Biomacromolecules 2016; 17:3547-3557. [PMID: 27696822 DOI: 10.1021/acs.biomac.6b01056] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human mitochondrial dysfunction can lead to severe and often deadly diseases, for which there are no known cures. Although the targeted delivery of therapeutic gene to mitochondria is a promising approach to alleviate these disorders, gene carrier systems for the selective delivery of functional DNA into the mitochondria of living mammalian cells are currently unavailable. Here we rationally developed dual-domain peptides containing DNA-condensing/cell-penetrating/endosome-disruptive and mitochondria-targeting sequences. Secondary structures of the dual-domain peptides were analyzed, and variations in the physicochemical properties (stability, size, and ζ potential) of peptide/DNA complexes were studied as a function of peptide-to-DNA ratio and serum addition. An optimized formulation, identified through qualitative and quantitative studies, fulfills the fundamental prerequisites for mitochondria-specific DNA delivery, successfully transfecting a high proportion (82 ± 2%) of mitochondria in a human cell line with concomitant biocompatibility. Nuclear magnetic resonance studies confirmed the effectiveness of our bipartite peptide design with segregated functions: a helical domain necessary for mitochondrial import and an unstructured region for interaction with DNA involving lysine residues. Further analyses revealed that the lysine-specific interaction assisted the self-organization of the peptide and the DNA cargo, leading to a structural arrangement within the formed complex that is crucial for its biological efficiency. Thus the reported gene vector represents a new and reliable tool to uncover the complexity of mitochondrial transfection.
Collapse
Affiliation(s)
- Jo-Ann Chuah
- Enzyme Research Team, Biomass Engineering Research Division, Center for Sustainable Resource Science, RIKEN , 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Akimasa Matsugami
- Systems and Structural Biology Center, Yokohama Institute, RIKEN , Tsurumi, 1-7-22 Suehiro-cho, Tsurumi-ku, Kanagawa 230-0045, Japan
| | - Fumiaki Hayashi
- Systems and Structural Biology Center, Yokohama Institute, RIKEN , Tsurumi, 1-7-22 Suehiro-cho, Tsurumi-ku, Kanagawa 230-0045, Japan
| | - Keiji Numata
- Enzyme Research Team, Biomass Engineering Research Division, Center for Sustainable Resource Science, RIKEN , 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| |
Collapse
|
126
|
Wang M, Liu H, Zheng J, Chen B, Zhou M, Fan W, Wang H, Liang X, Zhou X, Eriani G, Jiang P, Guan MX. A Deafness- and Diabetes-associated tRNA Mutation Causes Deficient Pseudouridinylation at Position 55 in tRNAGlu and Mitochondrial Dysfunction. J Biol Chem 2016; 291:21029-21041. [PMID: 27519417 DOI: 10.1074/jbc.m116.739482] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Indexed: 02/03/2023] Open
Abstract
Several mitochondrial tRNA mutations have been associated with maternally inherited diabetes and deafness. However, the pathophysiology of these tRNA mutations remains poorly understood. In this report, we identified the novel homoplasmic 14692A→G mutation in the mitochondrial tRNAGlu gene among three Han Chinese families with maternally inherited diabetes and deafness. The m.14692A→G mutation affected a highly conserved uridine at position 55 of the TΨC loop of tRNAGlu The uridine is modified to pseudouridine (Ψ55), which plays an important role in the structure and function of this tRNA. Using lymphoblastoid cell lines derived from a Chinese family, we demonstrated that the m.14692A→G mutation caused loss of Ψ55 modification and increased angiogenin-mediated endonucleolytic cleavage in mutant tRNAGlu The destabilization of base-pairing (18A-Ψ55) caused by the m.14692A→G mutation perturbed the conformation and stability of tRNAGlu An approximately 65% decrease in the steady-state level of tRNAGlu was observed in mutant cells compared with control cells. A failure in tRNAGlu metabolism impaired mitochondrial translation, especially for polypeptides with a high proportion of glutamic acid codons such as ND1, ND6, and CO2 in mutant cells. An impairment of mitochondrial translation caused defective respiratory capacity, especially reducing the activities of complexes I and IV. Furthermore, marked decreases in the levels of mitochondrial ATP and membrane potential were observed in mutant cells. These mitochondrial dysfunctions caused an increasing production of reactive oxygen species in the mutant cells. Our findings may provide new insights into the pathophysiology of maternally inherited diabetes and deafness, which is primarily manifested by the deficient nucleotide modification of mitochondrial tRNAGlu.
Collapse
Affiliation(s)
- Meng Wang
- From the Division of Clinical Genetics and Genomics, Children's Hospital and the Institute of Genetics, Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310001
| | - Hao Liu
- the Institute of Genetics, Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310001
| | - Jing Zheng
- From the Division of Clinical Genetics and Genomics, Children's Hospital and the Institute of Genetics, Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310001
| | - Bobei Chen
- the Department of Otolaryngology, Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035, the Attardi Institute of Mitochondrial Biomedicine, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Mi Zhou
- From the Division of Clinical Genetics and Genomics, Children's Hospital and the Institute of Genetics, Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310001
| | - Wenlu Fan
- the Attardi Institute of Mitochondrial Biomedicine, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Hen Wang
- the Attardi Institute of Mitochondrial Biomedicine, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China 325035
| | - Xiaoyang Liang
- From the Division of Clinical Genetics and Genomics, Children's Hospital and the Institute of Genetics, Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310001
| | - Xiaolong Zhou
- the Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China 200031, and
| | - Gilbert Eriani
- the Architecture et Réactivité de l'ARN, Université de Strasbourg, CNRS, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, 67084 Strasbourg, France
| | - Pingping Jiang
- From the Division of Clinical Genetics and Genomics, Children's Hospital and the Institute of Genetics, Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310001
| | - Min-Xin Guan
- From the Division of Clinical Genetics and Genomics, Children's Hospital and the Institute of Genetics, Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310001, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, and Joining Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, Zhejiang, China 310058,
| |
Collapse
|
127
|
Bridges between mitochondrial oxidative stress, ER stress and mTOR signaling in pancreatic β cells. Cell Signal 2016; 28:1099-104. [DOI: 10.1016/j.cellsig.2016.05.007] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 02/06/2023]
|
128
|
Dvorakova V, Kolarova H, Magner M, Tesarova M, Hansikova H, Zeman J, Honzik T. The phenotypic spectrum of fifty Czech m.3243A>G carriers. Mol Genet Metab 2016; 118:288-95. [PMID: 27296531 DOI: 10.1016/j.ymgme.2016.06.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 06/04/2016] [Accepted: 06/05/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Mitochondrial myopathy, Encephalopathy, Lactic Acidosis and Stroke-like episodes syndrome (MELAS) is a common mitochondrial disorder with varying multisystemic clinical manifestation. We present a comprehensive clinical picture of 50 Czech m.3243A>G carriers with emphasis on the sequence of symptoms in symptomatic patients. RESULTS Symptoms developed in 33 patients (66%) and 17 carriers remained unaffected (34%). The age of onset varied from 1month to 47years of age, with juvenile presentation occurring in 53% of patients. Myopathy was the most common presenting symptom (18%), followed by CPEO/ptosis and hearing loss, with the latter also being the most common second symptom. Stroke-like episodes (SLE) occurred in fourteen patients, although never as a first symptom, and were frequently preceded by migraines (58%). Rhabdomyolysis developed in two patients. The second symptom appeared 5.0±8.3years (range 0-28years) after the first, and the interval between the second and third symptom was 2.0±6.0years (range 0-21years). Four of our patients remained monosymptomatic up to 12years of follow-up. The sequence of symptoms according to their time of manifestation was migraines, myopathy, seizures, CPEO/ptosis, SLE, hearing loss, and diabetes mellitus. The average age at death was 32.4±17.7years (range 9-60years) in the juvenile form and 44.0±12.7years (range 35-53years) in the adult form. Some patients with SLE harboured very low heteroplasmy levels in various tissues. No threshold for any organ dysfunction could be determined based on these levels. CONCLUSIONS Sufficient knowledge of the timeline of the natural course of MELAS syndrome may improve the prediction and management of symptoms in patients with this mitochondrial disease.
Collapse
Affiliation(s)
- V Dvorakova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague, General University Hospital in Prague, Czech Republic
| | - H Kolarova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague, General University Hospital in Prague, Czech Republic
| | - M Magner
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague, General University Hospital in Prague, Czech Republic
| | - M Tesarova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague, General University Hospital in Prague, Czech Republic
| | - H Hansikova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague, General University Hospital in Prague, Czech Republic
| | - J Zeman
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague, General University Hospital in Prague, Czech Republic
| | - T Honzik
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague, General University Hospital in Prague, Czech Republic.
| |
Collapse
|
129
|
Blaychfeld-Magnazi M, Zornitzki T, Ulman M, Madar Z, Knobler H. Early beta-cell dysfunction characterizes males with type 2 diabetes of Yemenite origin. Acta Diabetol 2016; 53:567-74. [PMID: 26873241 DOI: 10.1007/s00592-016-0838-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/11/2016] [Indexed: 12/23/2022]
Abstract
AIMS The aim of the current study was to characterize β-cell function, insulin sensitivity and line of inheritance in patients with recent-onset type 2 diabetes of Yemenite and non-Yemenite Jewish origin. METHODS A cohort study including 121 GAD negative diabetic patients, 59 of Yemenite and 62 of non-Yemenite origin, treated by diet ± oral antihyperglycemic monotherapy who underwent 180-min meal tolerance test (MMT). Based on MMT, indexes of insulin resistance and secretion were calculated. RESULTS There were no significant differences in age, sex, diabetes duration, BMI, HbA1c and lipid profile. A significant difference was found in family history of diabetes: 63 % of patients of Yemenite origin had maternal inheritance versus 35 % in the non-Yemenite origin (p < 0.001). Both indexes of β-cell function, the insulinogenic and the disposition indexes were significantly lower in patients of Yemenite origin compared with non-Yemenite origin (0.66 ± 0.4 vs. 0.93 ± 0.8, p = 0.04; 2.3 ± 1.8 vs. 3.3 ± 3.3, p = 0.04, respectively) with no difference in insulin sensitivity. When females and males were analyzed separately, the difference in maternal inheritance remained significant in both, but the difference in β-cell function indexes was observed only in males (p = 0.03, p = 0.01, respectively). CONCLUSIONS Males with recent-onset diabetes of Yemenite origin have a significant reduction of β-cell function and reduced ability to compensate for insulin resistance compared with diabetic males of non-Yemenite origin. Both males and females of Yemenite origin have a significantly higher maternal inheritance of diabetes. These data suggest different underlying mechanisms leading to early loss of β-cell in diabetic males of Yemenite origin.
Collapse
Affiliation(s)
- Moran Blaychfeld-Magnazi
- Diabetes Endocrinology and Metabolic Disease Institute, Kaplan Medical Center, Hebrew University Medical School of Jerusalem, Bilu street 1, 76100, Rehovot, Israel
- Institute of Biochemistry Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Taiba Zornitzki
- Diabetes Endocrinology and Metabolic Disease Institute, Kaplan Medical Center, Hebrew University Medical School of Jerusalem, Bilu street 1, 76100, Rehovot, Israel.
| | - Mira Ulman
- Endocrinology Laboratory, Kaplan Medical Center, Rehovot, Israel
| | - Zecharia Madar
- Institute of Biochemistry Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Hilla Knobler
- Diabetes Endocrinology and Metabolic Disease Institute, Kaplan Medical Center, Hebrew University Medical School of Jerusalem, Bilu street 1, 76100, Rehovot, Israel
| |
Collapse
|
130
|
Zhang F, Liu C, Wang L, Cao X, Wang YY, Yang JK. Antioxidant effect of angiotensin (1‑7) in the protection of pancreatic β cell function. Mol Med Rep 2016; 14:1963-9. [PMID: 27430410 PMCID: PMC4991744 DOI: 10.3892/mmr.2016.5514] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 03/22/2016] [Indexed: 12/28/2022] Open
Abstract
It is well known that the local renin-angiotensin system (RAS) is activated in the diabetic state, which results in an increase in the level of oxidative stress injury to pancreatic β cells. The angiotensin-converting enzyme 2 (ACE2)/angiotensin (1-7) [Ang (1-7)]/Mas axis is a negative regulator of the classical renin-angiotensin system. In order to investigate the antioxidant effect of Ang (1-7) on pancreatic β cells, INS-1 cells were cultured and oxidative stress was induced by treatment with H2O2. Glucose-stimulated insulin secretion (GSIS), the generation of reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and glucose-stimulated calcium (GSCa) responses in β cells were determined following treatment with Ang (1-7). It was observed that H2O2 significantly impaired the insulin secreting function, increased the production of ROS, and also decreased the levels of GSCa and MMP. Pre-treatment with Ang (1-7) alleviated these effects and treatment with A779 [antagonist of Ang (1-7)] prevented the effects of Ang (1-7). Based on these findings, it was concluded that Ang (1-7) can protect pancreatic β cells from oxidative injury and such protection can be blocked by its antagonist A779.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Chang Liu
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Lei Wang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Xi Cao
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Ying Ying Wang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Jin Kui Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| |
Collapse
|
131
|
Flannick J, Johansson S, Njølstad PR. Common and rare forms of diabetes mellitus: towards a continuum of diabetes subtypes. Nat Rev Endocrinol 2016; 12:394-406. [PMID: 27080136 DOI: 10.1038/nrendo.2016.50] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Insights into the genetic basis of type 2 diabetes mellitus (T2DM) have been difficult to discern, despite substantial research. More is known about rare forms of diabetes mellitus, several of which share clinical and genetic features with the common form of T2DM. In this Review, we discuss the extent to which the study of rare and low-frequency mutations in large populations has begun to bridge the gap between rare and common forms of diabetes mellitus. We hypothesize that the perceived division between these diseases might be due, in part, to the historical ascertainment bias of genetic studies, rather than a clear distinction between disease pathophysiologies. We also discuss possible implications of a new model for the genetic basis of diabetes mellitus subtypes, where the boundary between subtypes becomes blurred.
Collapse
Affiliation(s)
- Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
- Center for Human Genetic Research, Massachusetts General Hospital, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| | - Stefan Johansson
- K.G. Jebsen Center for Diabetes Research, The Department of Clinical Science, University of Bergen, Jonas Lies veg 87, N-5020 Bergen, Norway
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Jonas Lies veg 65, N-5021 Bergen, Norway
| | - Pål R Njølstad
- K.G. Jebsen Center for Diabetes Research, The Department of Clinical Science, University of Bergen, Jonas Lies veg 87, N-5020 Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Jonas Lies veg 65, N-5021 Bergen, Norway
| |
Collapse
|
132
|
Reinauer C, Meissner T, Roden M, Thon A, Holterhus PM, Haberland H, Binder E, Marg W, Bollow E, Holl R. Low prevalence of patients with mitochondrial disease in the German/Austrian DPV diabetes registry. Eur J Pediatr 2016; 175:613-22. [PMID: 26670026 DOI: 10.1007/s00431-015-2675-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/20/2015] [Accepted: 11/26/2015] [Indexed: 01/21/2023]
Abstract
UNLABELLED The aim of this study was to characterize the phenotype and treatment of young patients (manifestation <30 years) with diabetes of mitochondrial origin (DMO), based on the German/Austrian DPV (Diabetes Patienten Verlaufsdokumentation) registry. Only 13 (0.02 %) of all patients with diabetes in this cohort were identified with DMO, mainly due to the Kearns-Sayre (n = 5), Pearson (n = 3), or mitochondrial myopathy, encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome (n = 2). The onset of DMO (14.2, interquartile range (IQR) 7.1-16 years) was later than diabetes onset in individuals with T1D but earlier than in T2D. At manifestation, patients exhibited a mild elevation of blood glucose concentrations (251, IQR 178-299 mg/dl) without ketoacidosis. They had lower body mass index (BMI) values (-1.39 ± 0.28 kg/m(2)) than peers with T1D or T2D (p < 0.0001) and higher triglycerides (211, IQR 134-574 mg/dl) than in T1D (p = 0.04) while there was a high rate of dyslipidemia (86 %). Insulin requirements (0.58, IQR 0.37-0.90 U/kg/d) were between T1D and T2D while glucometabolic control (glycated hemoglobin A1c (HbA1c) 7.4 ± 0.52 %) in DMO was comparable to age-matched T2D and stable over a 5-year follow-up. CONCLUSION Primary mitochondrial disorders are a rare cause of juvenile diabetes and likely to be underdiagnosed. As there is clinical overlap with T1D and T2D, dyslipidemia and low body weight may help to identify further DMO cases. WHAT IS KNOWN • In adults diabetes of mitochondrial origin (DMO) is a rare cause of non-autoimmune diabetes, affecting about 0.8 % of diabetes cases. • Common features are a maternal family history of diabetes, hearing loss and neurological abnormalities. What is New: • In our juvenile cohort 0.02 % of diabetes patients (age < 30 years) were affected by DMO, while Kearns Sayre, MELAS and Pearson syndrome were the most frequent entities. • Juvenile DMO patients exhibited dyslipidemia, higher triglycerides and a lower BMI than peers with T1D or T2D, while some patients also showed retinal changes.
Collapse
Affiliation(s)
- Christina Reinauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Partners Düsseldorf and Ulm, Düsseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD e.V.), Partners Düsseldorf and Ulm, Düsseldorf, Germany.,Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University Düsseldorf, Leibniz Center for Diabetes Research, German Center for Diabetes Research (DZD), 40225, Düsseldorf, Germany
| | - Angelika Thon
- Department of Pediatric Pneumology, Allergology & Neonatology, Children's Hospital, Hannover Medical School, 30625, Hannover, Germany
| | - Paul-Martin Holterhus
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University Hospital Schleswig-Holstein, Christian-Albrechts University Kiel, 24118, Kiel, Germany
| | - Holger Haberland
- Hospital for Children and Adolescents, Sana Hospital Berlin Lichtenberg, 10365, Berlin, Germany
| | - Elisabeth Binder
- Department of Pediatrics, Medical University of Innsbruck, A 6020, Innsbruck, Austria
| | - Wolfgang Marg
- Center for Pediatrics and Adolescent Medicine, Bremen-Mitte Hospital, 28211, Bremen, Germany
| | - Esther Bollow
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, German Center for Diabetes Research (DZD), 89081, Ulm, Germany
| | - Reinhard Holl
- Institute of Epidemiology and Medical Biometry, ZIBMT, University of Ulm, German Center for Diabetes Research (DZD), 89081, Ulm, Germany
| |
Collapse
|
133
|
Brun T, Maechler P. Beta-cell mitochondrial carriers and the diabetogenic stress response. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2540-9. [PMID: 26979549 DOI: 10.1016/j.bbamcr.2016.03.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 01/09/2023]
Abstract
Mitochondria play a central role in pancreatic beta-cells by coupling metabolism of the secretagogue glucose to distal events of regulated insulin exocytosis. This process requires transports of both metabolites and nucleotides in and out of the mitochondria. The molecular identification of mitochondrial carriers and their respective contribution to beta-cell function have been uncovered only recently. In type 2 diabetes, mitochondrial dysfunction is an early event and may precipitate beta-cell loss. Under diabetogenic conditions, characterized by glucotoxicity and lipotoxicity, the expression profile of mitochondrial carriers is selectively modified. This review describes the role of mitochondrial carriers in beta-cells and the selective changes in response to glucolipotoxicity. In particular, we discuss the importance of the transfer of metabolites (pyruvate, citrate, malate, and glutamate) and nucleotides (ATP, NADH, NADPH) for beta-cell function and dysfunction. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.
Collapse
Affiliation(s)
- Thierry Brun
- Department of Cell Physiology and Metabolism, Faculty Diabetes Center, Geneva University Medical Centre, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, Faculty Diabetes Center, Geneva University Medical Centre, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland.
| |
Collapse
|
134
|
Cross Talk of Proteostasis and Mitostasis in Cellular Homeodynamics, Ageing, and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4587691. [PMID: 26977249 PMCID: PMC4763003 DOI: 10.1155/2016/4587691] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/24/2015] [Accepted: 12/31/2015] [Indexed: 12/26/2022]
Abstract
Mitochondria are highly dynamic organelles that provide essential metabolic functions and represent the major bioenergetic hub of eukaryotic cell. Therefore, maintenance of mitochondria activity is necessary for the proper cellular function and survival. To this end, several mechanisms that act at different levels and time points have been developed to ensure mitochondria quality control. An interconnected highly integrated system of mitochondrial and cytosolic chaperones and proteases along with the fission/fusion machinery represents the surveillance scaffold of mitostasis. Moreover, nonreversible mitochondrial damage targets the organelle to a specific autophagic removal, namely, mitophagy. Beyond the organelle dynamics, the constant interaction with the ubiquitin-proteasome-system (UPS) has become an emerging aspect of healthy mitochondria. Dysfunction of mitochondria and UPS increases with age and correlates with many age-related diseases including cancer and neurodegeneration. In this review, we discuss the functional cross talk of proteostasis and mitostasis in cellular homeodynamics and the impairment of mitochondrial quality control during ageing, cancer, and neurodegeneration.
Collapse
|
135
|
A Metabolic Signature of Mitochondrial Dysfunction Revealed through a Monogenic Form of Leigh Syndrome. Cell Rep 2015; 13:981-9. [PMID: 26565911 PMCID: PMC4644511 DOI: 10.1016/j.celrep.2015.09.054] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 07/13/2015] [Accepted: 09/18/2015] [Indexed: 11/20/2022] Open
Abstract
A decline in mitochondrial respiration represents the root cause of a large number of inborn errors of metabolism. It is also associated with common age-associated diseases and the aging process. To gain insight into the systemic, biochemical consequences of respiratory chain dysfunction, we performed a case-control, prospective metabolic profiling study in a genetically homogenous cohort of patients with Leigh syndrome French Canadian variant, a mitochondrial respiratory chain disease due to loss-of-function mutations in LRPPRC. We discovered 45 plasma and urinary analytes discriminating patients from controls, including classic markers of mitochondrial metabolic dysfunction (lactate and acylcarnitines), as well as unexpected markers of cardiometabolic risk (insulin and adiponectin), amino acid catabolism linked to NADH status (α-hydroxybutyrate), and NAD+ biosynthesis (kynurenine and 3-hydroxyanthranilic acid). Our study identifies systemic, metabolic pathway derangements that can lie downstream of primary mitochondrial lesions, with implications for understanding how the organelle contributes to rare and common diseases.
Collapse
|
136
|
El-Hattab AW, Adesina AM, Jones J, Scaglia F. MELAS syndrome: Clinical manifestations, pathogenesis, and treatment options. Mol Genet Metab 2015; 116:4-12. [PMID: 26095523 DOI: 10.1016/j.ymgme.2015.06.004] [Citation(s) in RCA: 387] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/14/2015] [Accepted: 06/14/2015] [Indexed: 12/13/2022]
Abstract
Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is one of the most frequent maternally inherited mitochondrial disorders. MELAS syndrome is a multi-organ disease with broad manifestations including stroke-like episodes, dementia, epilepsy, lactic acidemia, myopathy, recurrent headaches, hearing impairment, diabetes, and short stature. The most common mutation associated with MELAS syndrome is the m.3243A>G mutation in the MT-TL1 gene encoding the mitochondrial tRNA(Leu(UUR)). The m.3243A>G mutation results in impaired mitochondrial translation and protein synthesis including the mitochondrial electron transport chain complex subunits leading to impaired mitochondrial energy production. The inability of dysfunctional mitochondria to generate sufficient energy to meet the needs of various organs results in the multi-organ dysfunction observed in MELAS syndrome. Energy deficiency can also stimulate mitochondrial proliferation in the smooth muscle and endothelial cells of small blood vessels leading to angiopathy and impaired blood perfusion in the microvasculature of several organs. These events will contribute to the complications observed in MELAS syndrome particularly the stroke-like episodes. In addition, nitric oxide deficiency occurs in MELAS syndrome and can contribute to its complications. There is no specific consensus approach for treating MELAS syndrome. Management is largely symptomatic and should involve a multidisciplinary team. Unblinded studies showed that l-arginine therapy improves stroke-like episode symptoms and decreases the frequency and severity of these episodes. Additionally, carnitine and coenzyme Q10 are commonly used in MELAS syndrome without proven efficacy.
Collapse
Affiliation(s)
- Ayman W El-Hattab
- Division of Clinical Genetics and Metabolic Disorders, Department of Pediatrics, Tawam Hospital, Al-Ain, United Arab Emirates
| | - Adekunle M Adesina
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Jeremy Jones
- Singleton Department of Radiology, Texas Children's Hospital, Houston, TX, USA
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
137
|
Lee J, Liu R, de Jesus D, Kim BS, Ma K, Moulik M, Yechoor V. Circadian control of β-cell function and stress responses. Diabetes Obes Metab 2015; 17 Suppl 1:123-33. [PMID: 26332977 PMCID: PMC4762487 DOI: 10.1111/dom.12524] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/27/2015] [Indexed: 12/20/2022]
Abstract
Circadian disruption is the bane of modern existence and its deleterious effects on health; in particular, diabetes and metabolic syndrome have been well recognized in shift workers. Recent human studies strongly implicate a 'dose-dependent' relationship between circadian disruption and diabetes. Genetic and environmental disruption of the circadian clock in rodents leads to diabetes secondary to β-cell failure. Deletion of Bmal1, a non-redundant core clock gene, leads to defects in β-cell stimulus-secretion coupling, decreased glucose-stimulated ATP production, uncoupling of OXPHOS and impaired glucose-stimulated insulin secretion. Both genetic and environmental circadian disruptions are sufficient to induce oxidative stress and this is mediated by a disruption of the direct transcriptional control of the core molecular clock and Bmal1 on Nrf2, the master antioxidant transcription factor in the β-cell. In addition, circadian disruption also leads to a dysregulation of the unfolded protein response and leads to endoplasmic reticulum stress in β-cells. Both the oxidative and endoplasmic reticulum (ER) stress contribute to an impairment of mitochondrial function and β-cell failure. Understanding the basis of the circadian control of these adaptive stress responses offers hope to target them for pharmacological modulation to prevent and mitigate the deleterious metabolic consequences of circadian disruption.
Collapse
Affiliation(s)
- J Lee
- Diabetes Research Center & Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston Texas USA 77030
| | - R Liu
- Diabetes Research Center & Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston Texas USA 77030
| | - D de Jesus
- Diabetes Research Center & Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston Texas USA 77030
| | - BS Kim
- Diabetes Research Center & Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston Texas USA 77030
| | - K Ma
- Center for Diabetes Research, The Methodist Hospital Research Institute, Houston Texas USA 77030
| | - M Moulik
- Division of Cardiology, Department of Pediatrics, University of Texas Medical School at Houston, Houston Texas USA 77030
| | - V Yechoor
- Diabetes Research Center & Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Baylor College of Medicine, Houston Texas USA 77030
- Dept of Molecular & Cellular Biology, Baylor College of Medicine; Houston Texas USA 77030
- Corresponding Author: Vijay Yechoor, MD, R612, One Baylor Plaza, Baylor College of Medicine, DERC & Division of Diabetes, Endocrinology & Metabolism, Department of Medicine, Houston TX 77030, Phone: 713-798-4146; Fax: 713-798-8764,
| |
Collapse
|
138
|
Liu B, Czajka A, Malik AN, Hussain K, Jones PM, Persaud SJ. Equilibrative nucleoside transporter 3 depletion in β-cells impairs mitochondrial function and promotes apoptosis: Relationship to pigmented hypertrichotic dermatosis with insulin-dependent diabetes. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2086-95. [PMID: 26163994 DOI: 10.1016/j.bbadis.2015.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/17/2015] [Accepted: 07/07/2015] [Indexed: 02/01/2023]
Abstract
Loss of function recessive mutations in the SLC29A3 gene that encodes human equilibrative nucleoside transporter 3 (ENT3) have been identified in patients with pigmented hypertrichotic dermatosis with insulin-dependent diabetes (PHID). ENT3 is a member of the equilibrative nucleoside transporter (ENT) family whose primary function is mediating transport of nucleosides and nucleobases. The aims of this study were to characterise ENT3 expression in islet β-cells and identify the effects of its depletion on β-cell mitochondrial activity and apoptosis. RT-PCR amplification identified ENT3 expression in human and mouse islets and exocrine pancreas, and in MIN6 β-cells. Immunohistochemistry using human and mouse pancreas sections exhibited extensive ENT3 immunostaining of β-cells, which was confirmed by co-staining with an anti-insulin antibody. In addition, exposure of dispersed human islet cells and MIN6 β-cells to MitoTracker and an ENT3 antibody showed co-localisation of ENT3 to β-cell mitochondria. Consistent with this, Western blot analysis confirmed enhanced ENT3 immunoreactivity in β-cell mitochondria-enriched fractions. Furthermore, ENT3 depletion in β-cells increased mitochondrial DNA content and promoted an energy crisis characterised by enhanced ATP-linked respiration and proton leak. Finally, inhibition of ENT3 activity by dypridamole and depletion of ENT3 by siRNA-induced knockdown resulted in increased caspase 3/7 activities in β-cells. These observations demonstrate that ENT3 is predominantly expressed by islet β-cells where it co-localises with mitochondria. Depletion of ENT3 causes mitochondrial dysfunction which is associated with enhanced β-cell apoptosis. Thus, apoptotic loss of islet β-cells may contribute to the occurrence of autoantibody-negative insulin-dependent diabetes in individuals with non-functional ENT3 mutations.
Collapse
Affiliation(s)
- B Liu
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life and Medical Sciences, King's College London, London SE1 1UL, United Kingdom
| | - A Czajka
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life and Medical Sciences, King's College London, London SE1 1UL, United Kingdom
| | - A N Malik
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life and Medical Sciences, King's College London, London SE1 1UL, United Kingdom
| | - K Hussain
- Institute of Child Health, London WC1N 1EH, United Kingdom
| | - P M Jones
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life and Medical Sciences, King's College London, London SE1 1UL, United Kingdom
| | - S J Persaud
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, Faculty of Life and Medical Sciences, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
139
|
Li W, Wen C, Li W, Wang H, Guan X, Zhang W, Ye W, Lu J. The tRNA(Gly) T10003C mutation in mitochondrial haplogroup M11b in a Chinese family with diabetes decreases the steady-state level of tRNA(Gly), increases aberrant reactive oxygen species production, and reduces mitochondrial membrane potential. Mol Cell Biochem 2015; 408:171-9. [PMID: 26134044 DOI: 10.1007/s11010-015-2493-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 06/18/2015] [Indexed: 11/28/2022]
Abstract
Mitochondrial diabetes originates mainly from mutations located in maternally transmitted, mitochondrial tRNA-coding genes. In a genetic screening program of type 2 diabetes conducted with a Chinese Han population, we found one family with suggestive maternally transmitted diabetes. The proband's mitochondrial genome was analyzed using DNA sequencing. Total 42 known nucleoside changes and 1 novel variant were identified, and the entire mitochondrial DNA sequence was assigned to haplogroup M11b. Phylogenetic analysis showed that a homoplasmic mutation, 10003T>C transition, occurred at the highly conserved site in the gene encoding tRNA(Gly). Using a transmitochondrial cybrid cell line harboring this mutation, we observed that the steady-state level of tRNA(Gly) significantly affected and the amount of tRNA(Gly) decreased by 97%, production of reactive oxygen species was enhanced, and mitochondrial membrane potential, mtDNA copy number and cellular oxygen consumption rate were remarkably decreased compared with wild-type cybrid cells. The homoplasmic 10003T>C mutation in the mitochondrial tRNA(Gly) gene suggested to be as a pathogenesis-related mutation which might contribute to the maternal inherited diabetes in the Han Chinese family.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, Zhejiang, People's Republic of China.,Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou, 325035, Zhejiang, People's Republic of China.,Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Higher Education Park, Chashan Town, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Chaowei Wen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Weixing Li
- Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Hailing Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Xiaomin Guan
- Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Wanlin Zhang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Wei Ye
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Jianxin Lu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Wenzhou, 325035, Zhejiang, People's Republic of China. .,Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou, 325035, Zhejiang, People's Republic of China. .,Wenzhou Medical University School of Laboratory Medicine and Life Sciences, Higher Education Park, Chashan Town, Wenzhou, 325035, Zhejiang, People's Republic of China.
| |
Collapse
|
140
|
Chen YT, Lin WD, Liao WL, Lin YJ, Chang JG, Tsai FJ. PTPRD silencing by DNA hypermethylation decreases insulin receptor signaling and leads to type 2 diabetes. Oncotarget 2015; 6:12997-3005. [PMID: 26079428 PMCID: PMC4536994 DOI: 10.18632/oncotarget.4092] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/11/2015] [Indexed: 11/28/2022] Open
Abstract
Genome-wide association study (GWAS) data showed that the protein tyrosine phosphatase receptor type delta (PTPRD) is associated with increased susceptibility to type 2 diabetes (T2D) in Han Chinese. A replication study indicated that PTPRD is involved in the insulin signaling pathway; however, the underlying mechanism remains unclear. We evaluated PTPRD expression in patients with T2D and controls. PTPRD expression levels were lower in patients and were correlated with the duration of the disease. Overexpression of the human insulin receptor PPARγ2 in HepG2 cells induced overexpression of PTPRD and the insulin receptor. PTPRD knockdown, using a shRNA, resulted in down-regulation of the insulin receptor. These results indicate that PTPRD activates PPARγ2 in the insulin signaling pathway. Similar results for PTPRD expression were found using a T2D mouse model. Silencing of PTPRD was caused by DNA methylation in T2D mice and patients, and correlated with DNMT1 expression. Furthermore, we showed that a DNMT1 SNP (rs78789647) was correlated with susceptibility to T2D. This study shows for the first time that DNMT1 caused PTPRD DNA hypermethylation and induced insulin signaling silencing in T2D patients. Our findings contribute to a better understanding of the crucial roles of these regulatory elements in human T2D.
Collapse
Affiliation(s)
- Yng-Tay Chen
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wei-De Lin
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Lin Liao
- Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Ying-Ju Lin
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Graduate Institute of China Medical Science, China Medical University, Taichung, Taiwan
| | - Jan-Gowth Chang
- Epigenome Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
141
|
Tabebi M, Mkaouar-Rebai E, Mnif M, Kallabi F, Ben Mahmoud A, Ben Saad W, Charfi N, Keskes-Ammar L, Kamoun H, Abid M, Fakhfakh F. A novel mutation MT-COIII m.9267G>C and MT-COI m.5913G>A mutation in mitochondrial genes in a Tunisian family with maternally inherited diabetes and deafness (MIDD) associated with severe nephropathy. Biochem Biophys Res Commun 2015; 459:353-60. [PMID: 25701779 DOI: 10.1016/j.bbrc.2015.01.151] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 01/29/2015] [Indexed: 11/17/2022]
Abstract
Mitochondrial diabetes (MD) is a heterogeneous disorder characterized by a chronic hyperglycemia, maternal transmission and its association with a bilateral hearing impairment. Several studies reported mutations in mitochondrial genes as potentially pathogenic for diabetes, since mitochondrial oxidative phosphorylation plays an important role in glucose-stimulated insulin secretion from beta cells. In the present report, we studied a Tunisian family with mitochondrial diabetes (MD) and deafness associated with nephropathy. The mutational analysis screening revealed the presence of a novel heteroplasmic mutation m.9276G>C in the mitochondrial COIII gene, detected in mtDNA extracted from leukocytes of a mother and her two daughters indicating that this mutation is maternally transmitted and suggest its implication in the observed phenotype. Bioinformatic tools showed that m.9267G>C mutation (p.A21P) is « deleterious » and it can modify the function and the stability of the MT-COIII protein by affecting the assembly of mitochondrial COX subunits and the translocation of protons then reducing the activity of the respective OXPHOS complexes of ATP synthesis. The nonsynonymous mutation (p.A21P) has not been reported before, it is the first mutation described in the COXIII gene which is related to insulin dependent mitochondrial diabetes and deafness and could be specific to the Tunisian population. The m.9267G>C mutation was present with a nonsynonymous inherited mitochondrial homoplasmic variation MT-COI m.5913 G>A (D4N) responsible of high blood pressure, a clinical feature detected in all explored patients.
Collapse
MESH Headings
- Adult
- Amino Acid Sequence
- Amino Acid Substitution
- Base Sequence
- Case-Control Studies
- Child, Preschool
- DNA Mutational Analysis
- DNA, Mitochondrial/genetics
- Deafness/complications
- Deafness/enzymology
- Deafness/genetics
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Electron Transport Complex IV/chemistry
- Electron Transport Complex IV/genetics
- Female
- Genes, Mitochondrial
- Humans
- Hypertension/complications
- Hypertension/enzymology
- Hypertension/genetics
- Kidney Diseases/complications
- Kidney Diseases/enzymology
- Kidney Diseases/genetics
- Male
- Middle Aged
- Mitochondrial Diseases
- Models, Molecular
- Molecular Sequence Data
- Mutation, Missense
- Pedigree
- Protein Structure, Secondary
- Sequence Homology, Amino Acid
- Tunisia
- Young Adult
Collapse
Affiliation(s)
- Mouna Tabebi
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Université de Sfax, Tunisia.
| | - Emna Mkaouar-Rebai
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Université de Sfax, Tunisia
| | - Mouna Mnif
- Service d'endocrinologie, C.H.U. Habib Bourguiba de Sfax, Tunisia
| | - Fakhri Kallabi
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Université de Sfax, Tunisia
| | - Afif Ben Mahmoud
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Université de Sfax, Tunisia
| | - Wafa Ben Saad
- Service d'endocrinologie, C.H.U. Habib Bourguiba de Sfax, Tunisia
| | - Nadia Charfi
- Service d'endocrinologie, C.H.U. Habib Bourguiba de Sfax, Tunisia
| | - Leila Keskes-Ammar
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Université de Sfax, Tunisia
| | - Hassen Kamoun
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Université de Sfax, Tunisia
| | - Mohamed Abid
- Service d'endocrinologie, C.H.U. Habib Bourguiba de Sfax, Tunisia
| | - Faiza Fakhfakh
- Laboratoire de Génétique Moléculaire Humaine, Faculté de Médecine de Sfax, Université de Sfax, Tunisia.
| |
Collapse
|
142
|
Shi M, Zheng J, Tan Y, Tan G, Li J, Li Y, Li X, Zhou Z, Yang R. Ultrasensitive Detection of Single Nucleotide Polymorphism in Human Mitochondrial DNA Utilizing Ion-Mediated Cascade Surface-Enhanced Raman Spectroscopy Amplification. Anal Chem 2015; 87:2734-40. [DOI: 10.1021/ac504000p] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Muling Shi
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Jing Zheng
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Yongjun Tan
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Guixiang Tan
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Jishan Li
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Yinhui Li
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Xia Li
- Xiangya Second Hospital of Central South University, Changsha, 410082, China
| | - Zhiguang Zhou
- Xiangya Second Hospital of Central South University, Changsha, 410082, China
| | - Ronghua Yang
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, College of Biology, Hunan University, Changsha, 410082, China
- School
of Chemistry and Biological Engineering, Changsha University of Science and Technology, Changsha, 410004, China
| |
Collapse
|
143
|
Brouwers MCGJ, Ham JC, Wisse E, Misra S, Landewe S, Rosenthal M, Patel D, Oliver N, Bilo HJG, Murphy E. Elevated lactate levels in patients with poorly regulated type 1 diabetes and glycogenic hepatopathy: a new feature of Mauriac syndrome. Diabetes Care 2015; 38:e11-2. [PMID: 25614691 DOI: 10.2337/dc14-2205] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Martijn C G J Brouwers
- Department of Endocrinology and Metabolic Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Janneke C Ham
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Eddie Wisse
- Electron Microscope Unit, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Shivani Misra
- Faculty of Medicine, Imperial College London, London, U.K
| | - Sabine Landewe
- Department of Endocrinology and Metabolic Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Miranda Rosenthal
- Department of Diabetes and Endocrinology, Royal Free London National Health Service Foundation Trust, London, U.K
| | - Dipesh Patel
- Department of Diabetes and Endocrinology, Royal Free London National Health Service Foundation Trust, London, U.K
| | - Nick Oliver
- Faculty of Medicine, Imperial College London, London, U.K
| | - Henk J G Bilo
- Department of Internal Medicine, Isala Klinieken Zwolle, Zwolle, the Netherlands
| | - Elaine Murphy
- Charles Dent Metabolic Unit, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, U.K
| |
Collapse
|
144
|
Maternally inherited diabetes is associated with a homoplasmic T10003C mutation in the mitochondrial tRNA(Gly) gene. Mitochondrion 2015; 21:49-57. [PMID: 25615420 DOI: 10.1016/j.mito.2015.01.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 12/27/2022]
Abstract
In this report, we investigate molecular pathogenic mechanism of a diabetes-associated homoplasmic mitochondrial tRNA mutation in a Han Chinese family with maternally transmitted diabetes mellitus. Of 10 adult matrilineal relatives, 5 individuals suffered from diabetes (4 subjects with only diabetes, one subject with both diabetes and hearing impairment), while other five matrilineal relatives (one with hearing loss) had glucose intolerance. The average age at onset of diabetes in matrilineal relatives was 50 years. Molecular analysis of their mitochondrial genomes identified the novel homoplasmic T10003C mutation in the tRNA(Gly) gene belonging to haplogroup M11b. The T10003C mutation is expected to form a base-pairing (13C-22G) at the highly conserved D-stem of tRNA(Gly), thereby affecting secondary structure and function of this tRNA. A tRNA Northern analysis revealed that the T10003C mutation caused ~70% reduction in the steady-state level of tRNA(Gly). An in vivo translation analysis showed ~33% reduction in the rate of mitochondrial translation in mutant cells. Oxygen consumption analysis showed the defects in overall respiratory capacity or the ATP-linked, proton leak, and maximal respiration in mutant cells. As a result, the cellular energy deficiency contributes to the development of diabetes in subjects carrying the T10003C mutation. These data provide the first direct evidence that the tRNA(Gly) mutation might be associated with diabetes. Thus, our findings may provide new insights into the understanding of pathophysiology of maternally inherited diabetes.
Collapse
|
145
|
Abstract
This article highlights the difficulties in creating a definitive classification of diabetes mellitus in the absence of a complete understanding of the pathogenesis of the major forms. This brief review shows the evolving nature of the classification of diabetes mellitus. No classification scheme is ideal, and all have some overlap and inconsistencies. The only diabetes in which it is possible to accurately diagnose by DNA sequencing, monogenic diabetes, remains undiagnosed in more than 90% of the individuals who have diabetes caused by one of the known gene mutations. The point of classification, or taxonomy, of disease, should be to give insight into both pathogenesis and treatment. It remains a source of frustration that all schemes of diabetes mellitus continue to fall short of this goal.
Collapse
Affiliation(s)
- Celeste C Thomas
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago, 5841 South Maryland Avenue, MC 1027, Chicago, IL 60637, USA.
| | - Louis H Philipson
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago, 5841 South Maryland Avenue, MC 1027, Chicago, IL 60637, USA; Department of Pediatrics, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago, 900 East 57th Street, Chicago, IL 60637, USA
| |
Collapse
|
146
|
Karalliedde J, Gnudi L. Diabetes mellitus, a complex and heterogeneous disease, and the role of insulin resistance as a determinant of diabetic kidney disease. Nephrol Dial Transplant 2014; 31:206-13. [PMID: 25550448 DOI: 10.1093/ndt/gfu405] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/02/2014] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is increasingly recognized as a heterogeneous condition. The individualization of care and treatment necessitates an understanding of the individual patient's pathophysiology of DM that underpins their DM classification and clinical presentation. Classical type-2 diabetes mellitus is due to a combination of insulin resistance and an insulin secretory defect. Type-1 diabetes is characterized by a near-absolute deficiency of insulin secretion. More recently, advances in genetics and a better appreciation of the atypical features of DM has resulted in more categories of diabetes. In the context of kidney disease, patients with DM and microalbuminuria are more insulin resistant, and insulin resistance may be a pathway that results in accelerated progression of diabetic kidney disease. This review summarizes the updated classification of DM, including more rarer categories and their associated renal manifestations that need to be considered in patients who present with atypical features. The benefits and limitations of the tests utilized to make a diagnosis of DM are discussed. We also review the putative pathways and mechanisms by which insulin resistance drives the progression of diabetic kidney disease.
Collapse
Affiliation(s)
- Janaka Karalliedde
- Unit for Metabolic Medicine, Department of Diabetes and Endocrinology, Cardiovascular Division, School of Life Science & Medicine, King's College, London, UK
| | - Luigi Gnudi
- Unit for Metabolic Medicine, Department of Diabetes and Endocrinology, Cardiovascular Division, School of Life Science & Medicine, King's College, London, UK
| |
Collapse
|
147
|
Nile DL, Brown AE, Kumaheri MA, Blair HR, Heggie A, Miwa S, Cree LM, Payne B, Chinnery PF, Brown L, Gunn DA, Walker M. Age-related mitochondrial DNA depletion and the impact on pancreatic Beta cell function. PLoS One 2014; 9:e115433. [PMID: 25532126 PMCID: PMC4274008 DOI: 10.1371/journal.pone.0115433] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/24/2014] [Indexed: 01/21/2023] Open
Abstract
Type 2 diabetes is characterised by an age-related decline in insulin secretion. We previously identified a 50% age-related decline in mitochondrial DNA (mtDNA) copy number in isolated human islets. The purpose of this study was to mimic this degree of mtDNA depletion in MIN6 cells to determine whether there is a direct impact on insulin secretion. Transcriptional silencing of mitochondrial transcription factor A, TFAM, decreased mtDNA levels by 40% in MIN6 cells. This level of mtDNA depletion significantly decreased mtDNA gene transcription and translation, resulting in reduced mitochondrial respiratory capacity and ATP production. Glucose-stimulated insulin secretion was impaired following partial mtDNA depletion, but was normalised following treatment with glibenclamide. This confirms that the deficit in the insulin secretory pathway precedes K+ channel closure, indicating that the impact of mtDNA depletion is at the level of mitochondrial respiration. In conclusion, partial mtDNA depletion to a degree comparable to that seen in aged human islets impaired mitochondrial function and directly decreased insulin secretion. Using our model of partial mtDNA depletion following targeted gene silencing of TFAM, we have managed to mimic the degree of mtDNA depletion observed in aged human islets, and have shown how this correlates with impaired insulin secretion. We therefore predict that the age-related mtDNA depletion in human islets is not simply a biomarker of the aging process, but will contribute to the age-related risk of type 2 diabetes.
Collapse
Affiliation(s)
- Donna L. Nile
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Audrey E. Brown
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Meutia A. Kumaheri
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Helen R. Blair
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Alison Heggie
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
| | - Satomi Miwa
- Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, NE4 5PL, United Kingdom
| | - Lynsey M. Cree
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Brendan Payne
- Wellcome Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, United Kingdom
| | - Patrick F. Chinnery
- Wellcome Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, United Kingdom
| | - Louise Brown
- Unilever R&D, Colworth Science Park, Sharnbrook, Bedford, MK44 1LQ, United Kingdom
| | - David A. Gunn
- Unilever R&D, Colworth Science Park, Sharnbrook, Bedford, MK44 1LQ, United Kingdom
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
- * E-mail:
| |
Collapse
|
148
|
Priyadarsini S, Sarker-Nag A, Allegood J, Chalfant C, Karamichos D. Description of the sphingolipid content and subspecies in the diabetic cornea. Curr Eye Res 2014; 40:1204-10. [PMID: 25426847 PMCID: PMC4763931 DOI: 10.3109/02713683.2014.990984] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Diabetes mellitus (DM) is characterized by high blood sugar levels over a prolonged period. Long term complications include but not limited heart disease, stroke, kidney failure, and ocular damage. An estimated 382 million people are diagnosed with Type 2 DM accounting for 90% of the cases. Common corneal dysfunctions associated with DM result in impaired vision due to decreased wound healing, corneal edema, and altered epithelial basement membrane. Lipids play a fundamental role in tissue metabolism and disease states. We attempt to determine the role of sphingolipids (SPL) in human Type I and Type II diabetic corneas. MATERIALS AND METHODS Cadaver corneas from healthy (non-diabetic/no ocular trauma), Type I (T1DM), and Type II diabetic (T2DM) donors were obtained and processed for lipidomics using LC-MS/MS. RESULTS Our data show significant differences in the SPL composition between control, T1DM and T2DM corneas. Both T1DM and T2DM showed a 10-folddownregulation of sphingomyelin(SM), 5-fold up regulation of Ceramides (Cer) and 2-fold upregulation of monohexosylceramides (MHC). Differences were also seen in total amounts of SPL where Cer was increased by approximately 3 fold in both T1DM and T2DM where SM decreased by 50% in both T1DM and T2DM when compared to healthy controls. No differences were seen in MHC amounts. CONCLUSIONS Overall, our data indicate major differences in SPL distribution in human diabetic corneas. Information on the sphingolipids role in cornea, corneal cell physiology, and diseases are very limitedwhich highlights the importance of these findings.
Collapse
Affiliation(s)
- Shrestha Priyadarsini
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Akhee Sarker-Nag
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jeremy Allegood
- Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA 23249
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University-School of Medicine, Richmond, VA 23298-0614
- The VCU Johnson Center, Richmond, VA 23298
- The VCU Massey Cancer Center, Richmond, VA 23298
| | - Charles Chalfant
- Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA 23249
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University-School of Medicine, Richmond, VA 23298-0614
- The VCU Johnson Center, Richmond, VA 23298
- The VCU Massey Cancer Center, Richmond, VA 23298
| | - Dimitrios Karamichos
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
149
|
Adrenal insufficiency in a child with MELAS syndrome. Brain Dev 2014; 36:924-7. [PMID: 24508408 DOI: 10.1016/j.braindev.2013.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 12/28/2013] [Accepted: 12/31/2013] [Indexed: 11/24/2022]
Abstract
Mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS) are established subgroups of mitochondrial encephalomyopathy. m.3243A>G a common point mutation is detected in tRNA in majority of patients with MELAS phenotype whereas m.8344A>G point mutation in tRNA is observed, in MERRF phenotype. Adrenal insufficiency has not been reported in mitochondrial disease, except in Kearns-Sayre Syndrome (KSS), which is a mitochondrial deletion syndrome. We report an unusual presentation in a five year old boy who presented with clinical phenotype of MELAS and was found to have m.8344A>G mutation in tRNA. Addison disease was identified due to hyperpigmentation of lips and gums present from early childhood. This is the first report describing adrenal insufficiency in a child with MELAS phenotype.
Collapse
|
150
|
Pal S, Sarkar C. Protective effect of resveratrol on fluoride induced alteration in protein and nucleic acid metabolism, DNA damage and biogenic amines in rat brain. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:684-699. [PMID: 25233527 DOI: 10.1016/j.etap.2014.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 06/03/2023]
Abstract
Fluoride, a well-established environmental carcinogen, has been found to cause various neurodegenerative diseases in human. Sub-acute exposure to fluoride at a dose of 20mg/kgb.w./day for 30 days caused significant alteration in pro-oxidant/anti-oxidant status of brain tissue as reflected by perturbation of reduced glutathione content, increased lipid peroxidation, protein carbonylation, nitric oxide and free hydroxyl radical production and decreased activities of antioxidant enzymes. Decreased proteolytic and transaminase enzymes' activities, protein and nucleic acid contents and associated DNA damage were observed in the brain of fluoride intoxicated rats. The neurotransmitters dopamine (DA), norepinephrine (NE) and serotonin level was also significantly altered after fluoride exposure. Protective effect of resveratrol on fluoride-induced metabolic and oxidative dysfunctions was evaluated. Resveratrol was found to inhibit changes in metabolic activities restoring antioxidant status, biogenic amine level and structural organization of the brain. Our findings indicated that resveratrol imparted antioxidative role in ameliorating fluoride-induced metabolic and oxidative stress in different regions of the brain.
Collapse
Affiliation(s)
- Sudipta Pal
- Nutritional Biochemistry Laboratory, Department of Human Physiology, Tripura University (A Central University), Suryamaninagar, West Tripura, Agartala 799022, India.
| | - Chaitali Sarkar
- Nutritional Biochemistry Laboratory, Department of Human Physiology, Tripura University (A Central University), Suryamaninagar, West Tripura, Agartala 799022, India
| |
Collapse
|