1
|
Pecina P, Čunátová K, Kaplanová V, Puertas-Frias G, Šilhavý J, Tauchmannová K, Vrbacký M, Čajka T, Gahura O, Hlaváčková M, Stránecký V, Kmoch S, Pravenec M, Houštěk J, Mráček T, Pecinová A. Haplotype variability in mitochondrial rRNA predisposes to metabolic syndrome. Commun Biol 2024; 7:1116. [PMID: 39261587 PMCID: PMC11391015 DOI: 10.1038/s42003-024-06819-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Metabolic syndrome is a growing concern in developed societies and due to its polygenic nature, the genetic component is only slowly being elucidated. Common mitochondrial DNA sequence variants have been associated with symptoms of metabolic syndrome and may, therefore, be relevant players in the genetics of metabolic syndrome. We investigate the effect of mitochondrial sequence variation on the metabolic phenotype in conplastic rat strains with identical nuclear but unique mitochondrial genomes, challenged by high-fat diet. We find that the variation in mitochondrial rRNA sequence represents risk factor in the insulin resistance development, which is associated with diacylglycerols accumulation, induced by tissue-specific reduction of the oxidative capacity. These metabolic perturbations stem from the 12S rRNA sequence variation affecting mitochondrial ribosome assembly and translation. Our work demonstrates that physiological variation in mitochondrial rRNA might represent a relevant underlying factor in the progression of metabolic syndrome.
Collapse
Affiliation(s)
- Petr Pecina
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kristýna Čunátová
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Vilma Kaplanová
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Guillermo Puertas-Frias
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Šilhavý
- Laboratory of Genetics of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kateřina Tauchmannová
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Marek Vrbacký
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Čajka
- Laboratory of Translational Metabolism, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Ondřej Gahura
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Markéta Hlaváčková
- Laboratory of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Viktor Stránecký
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michal Pravenec
- Laboratory of Genetics of Model Diseases, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Josef Houštěk
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Mráček
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| | - Alena Pecinová
- Laboratory of Bioenergetics, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
2
|
Chang X, Qu HQ, Liu Y, Glessner JT, Hakonarson H. Mitochondrial DNA Haplogroup K Is Protective Against Autism Spectrum Disorder Risk in Populations of European Ancestry. J Am Acad Child Adolesc Psychiatry 2024; 63:835-844. [PMID: 38072244 PMCID: PMC11186604 DOI: 10.1016/j.jaac.2023.09.550] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 09/23/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Accumulative evidence indicates a critical role of mitochondrial function in autism spectrum disorders (ASD), implying that ASD risk may be linked to mitochondrial dysfunction due to DNA (mtDNA) variations. Although a few studies have explored the association between mtDNA variations and ASD, the role of mtDNA in ASD is still unclear. Here, we aimed to investigate whether mitochondrial DNA haplogroups are associated with the risk of ASD. METHOD Two European cohorts and an Ashkenazi Jewish (AJ) cohort were analyzed, including 2,062 ASD patients in comparison with 4,632 healthy controls. DNA samples were genotyped using Illumina HumanHap550/610 and Illumina 1M arrays, inclusive of mitochondrial markers. Mitochondrial DNA (mtDNA) haplogroups were identified from genotyping data using HaploGrep2. A mitochondrial genome imputation pipeline was established to detect mtDNA variants. We conducted a case-control study to investigate potential associations of mtDNA haplogroups and variants with the susceptibility to ASD. RESULTS We observed that the ancient adaptive mtDNA haplogroup K was significantly associated with decreased risk of ASD by the investigation of 2 European cohorts including a total of 2,006 cases and 4,435 controls (odds ratio = 0.64, P=1.79 × 10-5), and we replicated this association in an Ashkenazi Jewish (AJ) cohort including 56 cases and 197 controls (odds ratio = 0.35, P = 9.46 × 10-3). Moreover, we demonstrate that the mtDNA variants rs28358571, rs28358584, and rs28358280 are significantly associated with ASD risk. Further expression quantitative trait loci (eQTLs) analysis indicated that the rs28358584 and rs28358280 genotypes are associated with expression levels of nearby genes in brain tissues, suggesting those mtDNA variants may confer risk for ASD via regulation of expression levels of genes encoded by the mitochondrial genome. CONCLUSION This study helps to shed light on the contribution of mitochondria in ASD and provides new insights into the genetic mechanism underlying ASD, suggesting the potential involvement of mtDNA-encoded proteins in the development of ASD. PLAIN LANGUAGE SUMMARY Increasing evidence indicates that mitochondrial dysfunction may be linked to autism spectrum disorder (ASD). This study investigated potential associations of mitochondrial DNA (mtDNA) variants in 2 European and Ashkenazi Jewish cohorts including 2,062 individuals with ASD and 4,632 healthy controls. Researchers found that the ancient mtDNA haplogroup K was linked to a reduced risk of ASD in both European and Ashkenazi Jewish populations. Additionally, specific mtDNA variants were associated with ASD risk and were shown to influence the expression of nearby genes in the brain. These findings highlight the potential involvement of mtDNA in ASD development, offering new insights into the genetic mechanisms underlying the disorder.
Collapse
Affiliation(s)
- Xiao Chang
- Children's Hospital of Philadelphia, Pennsylvania, United States; Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China.
| | - Hui-Qi Qu
- Children's Hospital of Philadelphia, Pennsylvania, United States
| | - Yichuan Liu
- Children's Hospital of Philadelphia, Pennsylvania, United States
| | | | - Hakon Hakonarson
- Children's Hospital of Philadelphia, Pennsylvania, United States; The Perelman School of Medicine, University of Pennsylvania, Pennsylvania, United States and Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
3
|
Guo H, Li P, Zhao J, Xin Q, Miao Y, Li L, Li X, Wang S, Mo H, Zeng L, Ju Z, Liu Z, Shen X, Cong W. Sheng Mai Yin shows anti-fatigue, anti-hypoxia and cardioprotective potential in an experimental joint model of fatigue and acute myocardial infarction. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117338. [PMID: 37890804 DOI: 10.1016/j.jep.2023.117338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiovascular disease (CVD) and fatigue are two common diseases endangering human life and health that may interact and reinforce one another. Myocardial infarction survivors frequently experience fatigue, and acute myocardial infarction (AMI) is one of the most common cardiovascular diseases that cause fatigue-induced sudden death. Sheng Mai Yin (SMY), a Chinese medicine prescription, is traditionally used for the treatment of diabetes and cardiovascular disease, and has been demonstrated to reduce fatigue and safeguard cardiac function. AIM OF THE STUDY This study aims to investigate the effects and underlying mechanisms of SMY in treating fatigue and AMI. MATERIALS AND METHODS The pharmacological mechanisms of SMY in treating fatigue and AMI were predicted by bioinformatics and network pharmacology methods. After administering SMY at high, medium and low doses, the swimming time to exhaustion, hemoglobin level, serological parameters and hypoxia tolerance time were detected in C57BL/6N mice, and the left ventricular ejection fractions (LVEF), left ventricular fractional shortening (LVFS), grasp strength, cardiac histopathology, serological parameters and the expression of PINK1 and Parkin proteins were examined in Wistar rats. RESULTS 371 core targets for SMY and 282 disease targets for fatigue and AMI were obtained using bioinformatics and network pharmacology methods. Enrichment analysis of target genes revealed that SMY might interfere with fatigue and AMI through biological processes such as mitochondrial autophagy, apoptosis, and oxidative stress. For in vivo experiments, SMY showed significant anti-fatigue and hypoxia tolerance effects in mice; It also improved the cardiac function and grasp strength, decreased their cardiac index, myocardial injury and fibrosis degree, and induced serological parameters levels and the expression of PTEN-induced putative kinase 1 (PINK1) and Parkin proteins in myocardium, suggesting that SMY may exert cardioprotective effects in a joint rat model of fatigue and AMI by inhibiting excessive mitochondrial autophagy. CONCLUSION This study revealed the anti-fatigue, anti-hypoxia and cardioprotective effects of SMY in a joint model of fatigue-AMI, and the pharmacological mechanism may be related to the inhibition of mitochondrial autophagy in cardiomyocytes through the PINK1/Parkin pathway. The discoveries may provide new ideas for the mechanism study of traditional Chinese medicine, especially complex prescriptions, in treating fatigue and AMI.
Collapse
Affiliation(s)
- Hao Guo
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China
| | - Jun Zhao
- Traditional Chinese Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China
| | - Li Li
- Chenland Research Institute, Irvine, CA, 92614, USA
| | - Xin Li
- Chenland Research Institute, Irvine, CA, 92614, USA
| | | | - Hui Mo
- Macao Health Bureau, Macao, 999078, China
| | - Li Zeng
- Macau University of Science and Technology, Macao, 999078, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Zimin Liu
- Chenland Research Institute, Irvine, CA, 92614, USA.
| | - Xiaoxu Shen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100091, China.
| | - Weihong Cong
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Beijing, 100091, China.
| |
Collapse
|
4
|
Byappanahalli AM, Omoniyi V, Noren Hooten N, Smith JT, Mode NA, Ezike N, Zonderman AB, Evans MK. Extracellular vesicle mitochondrial DNA levels are associated with race and mitochondrial DNA haplogroup. iScience 2024; 27:108724. [PMID: 38226163 PMCID: PMC10788249 DOI: 10.1016/j.isci.2023.108724] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/08/2023] [Accepted: 12/11/2023] [Indexed: 01/17/2024] Open
Abstract
Circulating cell-free mitochondrial DNA (ccf-mtDNA) acts as a damage-associated molecular pattern molecule and may be cargo within extracellular vesicles (EVs). ccf-mtDNA and select mitochondrial DNA (mtDNA) haplogroups are associated with cardiovascular disease. We hypothesized that ccf-mtDNA and plasma EV mtDNA would be associated with hypertension, sex, self-identified race, and mtDNA haplogroup ancestry. Participants were normotensive (n = 107) and hypertensive (n = 108) African American and White adults from the Healthy Aging in Neighborhoods of Diversity across the Life Span study. ccf-mtDNA levels were higher in African American participants compared with White participants in both plasma and EVs, but ccf-mtDNA levels were not related to hypertension. EV mtDNA levels were highest in African American participants with African mtDNA haplogroup. Circulating inflammatory protein levels were altered with mtDNA haplogroup, race, and EV mtDNA. Our findings highlight that race is a social construct and that ancestry is crucial when examining health and biomarker differences between groups.
Collapse
Affiliation(s)
- Anjali M. Byappanahalli
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Victor Omoniyi
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Jessica T. Smith
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Nicolle A. Mode
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Ngozi Ezike
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| |
Collapse
|
5
|
Onieva A, Martin J, R Cuesta-Aguirre D, Planells V, Coronado-Zamora M, Beyer K, Vega T, Lozano JE, Santos C, Aluja MP. Complete mitochondrial DNA profile in stroke: A geographical matched case-control study in Spanish population. Mitochondrion 2023; 73:51-61. [PMID: 37793469 DOI: 10.1016/j.mito.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/28/2023] [Accepted: 10/01/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION Stroke, the second leading cause of death worldwide, is a complex disease influenced by many risk factors among which we can find reactive oxygen species (ROS). Since mitochondria are the main producers of cellular ROS, nowadays studies are trying to elucidate the role of these organelles and its DNA (mtDNA) variation in stroke risk. The aim of the present study was to perform a comprehensive evaluation of the association between mtDNA mutations and mtDNA content and stroke risk. MATERIAL AND METHODS Homoplasmic and heteroplasmic mutations of the mtDNA were analysed in a case-controls study using 110 S cases and their corresponding control individuals. Mitochondrial DNA copy number (mtDNA-CN) was analysed in 73 of those case-control pairs. RESULTS Our results suggest that haplogroup V, specifically variants m.72C > T, m.4580G > A, m.15904C > T and m.16298 T > C have a protective role in relation to stroke risk. On the contrary, variants m.73A > G, m.11719G > A and m.14766C > T appear to be genetic risk factors for stroke. In this study, we found no statistically significant association between stroke risk and mitochondrial DNA copy number. CONCLUSIONS These results demonstrate the possible role of mtDNA genetics on the pathogenesis of stroke, probably through alterations in mitochondrial ROS production.
Collapse
Affiliation(s)
- Ana Onieva
- Unitat d'Antropologia Biològica, Departament BAVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Joan Martin
- Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Daniel R Cuesta-Aguirre
- Unitat d'Antropologia Biològica, Departament BAVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Violeta Planells
- Unitat d'Antropologia Biològica, Departament BAVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Marta Coronado-Zamora
- Institut de Biotecnologia i Biomedicina; Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Katrin Beyer
- Department of Pathology, Germans Trias i Pujol Research Institute, Badalona 08916 Barcelona, Spain
| | - Tomás Vega
- Dirección General de Salud Pública. Consejería de Sanidad. Junta de Castilla y León, 47007 Valladolid, Spain
| | - José Eugenio Lozano
- Dirección General de Salud Pública. Consejería de Sanidad. Junta de Castilla y León, 47007 Valladolid, Spain
| | - Cristina Santos
- Unitat d'Antropologia Biològica, Departament BAVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Maria Pilar Aluja
- Unitat d'Antropologia Biològica, Departament BAVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
6
|
O’Neill KN, Aubrey E, Howe LD, Stergiakouli E, Rodriguez S, Kearney PM, O’Keeffe LM. Mitochondrial DNA haplogroups and trajectories of cardiometabolic risk factors during childhood and adolescence: A prospective cohort study. PLoS One 2023; 18:e0284226. [PMID: 37043466 PMCID: PMC10096512 DOI: 10.1371/journal.pone.0284226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/27/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Mitochondria are organelles responsible for converting glucose into energy. Mitochondrial DNA is exclusively maternally inherited. The role of mitochondrial DNA haplogroups in the aetiology of cardiometabolic disease risk is not well understood. METHODS Sex-specific associations between common European mitochondrial DNA haplogroups (H, U, J, T, K, V, W, I and X) and trajectories of cardiometabolic risk factors from birth to 18 years were examined in a prospective cohort. Cardiometabolic risk factors measured from birth/mid-childhood to 18 years included body mass index (BMI), fat and lean mass, systolic and diastolic blood pressure, pulse rate, high-density lipoprotein cholesterol (HDL-c), non-HDL-c and triglycerides. Fractional polynomial and linear spline multilevel models explored the sex-specific association between haplogroups and risk factor trajectories. RESULTS Among a total of 7,954 participants with 79,178 repeated measures per outcome, we found no evidence that haplogroups U, T, J, K and W were associated with cardiometabolic risk factors compared to haplogroup H. In females, haplogroup V was associated with 4.0% (99% CI: -7.5, -0.6) lower BMI at age one but associations did not persist at age 18. Haplogroup X was associated with 1.3kg (99% CI: -2.5, -0.2) lower lean mass at age 9 which persisted at 18. Haplogroup V and X were associated with 9.3% (99% CI: -0.4, 19.0) and 16.4% (99% CI: -0.5,33.3) lower fat mass at age 9, respectively, although confidence intervals spanned the null and associations did not persist at 18. In males, haplogroup I was associated with 2.4% (99% CI: -0.5, 5.3) higher BMI at age 7; widening to 5.1% (99% CI: -0.5, 10.6) at 18 with confidence intervals spanning the null. CONCLUSIONS Our study demonstrated little evidence of sex-specific associations between mitochondrial DNA haplogroups and cardiometabolic risk factors.
Collapse
Affiliation(s)
- Kate N. O’Neill
- School of Public Health, University College Cork, Cork, Ireland
| | - Emily Aubrey
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| | - Laura D. Howe
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| | - Evie Stergiakouli
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| | - Santiago Rodriguez
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| | | | - Linda M. O’Keeffe
- School of Public Health, University College Cork, Cork, Ireland
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| |
Collapse
|
7
|
Kumar R, Harilal S, Thomas Parambi DG, Kanthlal S, Rahman MA, Alexiou A, Batiha GES, Mathew B. The Role of Mitochondrial Genes in Neurodegenerative Disorders. Curr Neuropharmacol 2022; 20:824-835. [PMID: 34503413 PMCID: PMC9881096 DOI: 10.2174/1570159x19666210908163839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/09/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial disorders are clinically heterogeneous, resulting from nuclear gene and mitochondrial mutations that disturb the mitochondrial functions and dynamics. There is a lack of evidence linking mtDNA mutations to neurodegenerative disorders, mainly due to the absence of noticeable neuropathological lesions in postmortem samples. This review describes various gene mutations in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, and stroke. These abnormalities, including PINK1, Parkin, and SOD1 mutations, seem to reveal mitochondrial dysfunctions due to either mtDNA mutation or deletion, the mechanism of which remains unclear in depth.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Della Grace Thomas Parambi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf-2014, Saudi Arabia
| | - S.K. Kanthlal
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, Hebersham, Australia;,AFNP Med Austria, Wien, Austria
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India,Address correspondence to this author at the Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi-682 041, India; E-mails: ;
| |
Collapse
|
8
|
Lee CS, Kim D, Hwang CS. Where Does N-Formylmethionine Come from? What for? Where Is It Going? What is the origin of N-formylmethionine in eukaryotic cells? Mol Cells 2022; 45:109-111. [PMID: 35288488 PMCID: PMC8926868 DOI: 10.14348/molcells.2021.5040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 11/27/2022] Open
Affiliation(s)
- Chang-Seok Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Dasom Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Cheol-Sang Hwang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
9
|
Nouri-Vaskeh M, Khalili N, Sadighi A, Yazdani Y, Zand R. Biomarkers for Transient Ischemic Attack: A Brief Perspective of Current Reports and Future Horizons. J Clin Med 2022; 11:jcm11041046. [PMID: 35207321 PMCID: PMC8877275 DOI: 10.3390/jcm11041046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 02/05/2023] Open
Abstract
Cerebrovascular disease is the leading cause of long-term disability in the world and the third-leading cause of death in the United States. The early diagnosis of transient ischemic attack (TIA) is of great importance for reducing the mortality and morbidity of cerebrovascular diseases. Patients with TIA have a high risk of early subsequent ischemic stroke and the development of permanent nervous system lesions. The diagnosis of TIA remains a clinical diagnosis that highly relies on the patient's medical history assessment. There is a growing list of biomarkers associated with different components of the ischemic cascade in the brain. In this review, we take a closer look at the biomarkers of TIA and their validity with a focus on the more clinically important ones using recent evidence of their reliability for practical usage.
Collapse
Affiliation(s)
- Masoud Nouri-Vaskeh
- Tropical and Communicable Diseases Research Centre, Iranshahr University of Medical Sciences, Iranshahr 7618815676, Iran;
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Tehran 1419733151, Iran
| | - Neda Khalili
- School of Medicine, Tehran University of Medical Sciences, Tehran 1449614535, Iran;
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Alireza Sadighi
- Neuroscience Institute, Geisinger Health System, Danville, PA 17822, USA;
| | - Yalda Yazdani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran;
| | - Ramin Zand
- Neuroscience Institute, Geisinger Health System, Danville, PA 17822, USA;
- Neuroscience Institute, Pennsylvania State University, State College, PA 16801, USA
- Correspondence: or ; Tel.: +1-570-808-7330; Fax: +1-570-808-3209
| |
Collapse
|
10
|
The role of mtDNA haplogroups on metabolic features in narcolepsy type 1. Mitochondrion 2022; 63:37-42. [PMID: 35051655 DOI: 10.1016/j.mito.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 11/24/2022]
Abstract
Narcolepsy type 1 (NT1) is due to selective loss of hypocretin (hcrt)-producing-neurons. Hcrt is a neuropeptide regulating the sleep/wake cycle, as well as feeding behavior. A subset of NT1 patients become overweight/obese, with a dysmetabolic phenotype. We hypothesized that mitochondrial DNA (mtDNA) sequence variation might contribute to the metabolic features in NT1 and we undertook an exploratory survey of mtDNA haplogroups in a cohort of well-characterized patients. We studied 246 NT1 Italian patients, fully defined for their metabolic features, including obesity, hypertension, low HDL, hypertriglyceridemia and hyperglycemia. For haplogroup assignment, the mtDNA control region was sequenced in combination with an assessment of diagnostic markers in the coding region. NT1 patients displayed the same mtDNA haplogroups (H, HV, J, K, T, U) frequency as those reported in the general Italian population. The majority of NT1 patients (64%) were overweight: amongst these, 35% were obese, 48% had low HDL cholesterol levels, and 31% had hypertriglyceridemia. We identified an association between haplogroups J, K and hypertriglyceridemia (P=0.03, 61.5% and 61.5%, respectively vs. 31.3% of the whole sample) and after correction for age and sex, we observed a reduction of these associations (OR=3.65, 95%CI=0.76-17.5, p=0.106 and 1.73, 0.52-5.69, p=0.368, respectively). The low HDL level showed a trend for association with haplogroup J (P=0.09, 83.3% vs. 47.4% of the whole sample) and after correction we observed an OR=6.73, 95%CI=0.65-69.9, p=0.110. Our study provides the first indication that mtDNA haplogroups J and K can modulate metabolic features of NT1 patients, linking mtDNA variation to the dysmetabolic phenotype in NT1.
Collapse
|
11
|
Singh LN, Kao SH, Wallace DC. Unlocking the Complexity of Mitochondrial DNA: A Key to Understanding Neurodegenerative Disease Caused by Injury. Cells 2021; 10:cells10123460. [PMID: 34943968 PMCID: PMC8715673 DOI: 10.3390/cells10123460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders that are triggered by injury typically have variable and unpredictable outcomes due to the complex and multifactorial cascade of events following the injury and during recovery. Hence, several factors beyond the initial injury likely contribute to the disease progression and pathology, and among these are genetic factors. Genetics is a recognized factor in determining the outcome of common neurodegenerative diseases. The role of mitochondrial genetics and function in traditional neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases, is well-established. Much less is known about mitochondrial genetics, however, regarding neurodegenerative diseases that result from injuries such as traumatic brain injury and ischaemic stroke. We discuss the potential role of mitochondrial DNA genetics in the progression and outcome of injury-related neurodegenerative diseases. We present a guide for understanding mitochondrial genetic variation, along with the nuances of quantifying mitochondrial DNA variation. Evidence supporting a role for mitochondrial DNA as a risk factor for neurodegenerative disease is also reviewed and examined. Further research into the impact of mitochondrial DNA on neurodegenerative disease resulting from injury will likely offer key insights into the genetic factors that determine the outcome of these diseases together with potential targets for treatment.
Collapse
Affiliation(s)
- Larry N. Singh
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Correspondence:
| | - Shih-Han Kao
- Resuscitation Science Center, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Department of Pediatrics, Division of Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Mehrabi M, Morris TA, Cang Z, Nguyen CHH, Sha Y, Asad MN, Khachikyan N, Greene TL, Becker DM, Nie Q, Zaragoza MV, Grosberg A. A Study of Gene Expression, Structure, and Contractility of iPSC-Derived Cardiac Myocytes from a Family with Heart Disease due to LMNA Mutation. Ann Biomed Eng 2021; 49:3524-3539. [PMID: 34585335 PMCID: PMC8671287 DOI: 10.1007/s10439-021-02850-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Genetic mutations to the Lamin A/C gene (LMNA) can cause heart disease, but the mechanisms making cardiac tissues uniquely vulnerable to the mutations remain largely unknown. Further, patients with LMNA mutations have highly variable presentation of heart disease progression and type. In vitro patient-specific experiments could provide a powerful platform for studying this phenomenon, but the use of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) introduces heterogeneity in maturity and function thus complicating the interpretation of the results of any single experiment. We hypothesized that integrating single cell RNA sequencing (scRNA-seq) with analysis of the tissue architecture and contractile function would elucidate some of the probable mechanisms. To test this, we investigated five iPSC-CM lines, three controls and two patients with a (c.357-2A>G) mutation. The patient iPSC-CM tissues had significantly weaker stress generation potential than control iPSC-CM tissues demonstrating the viability of our in vitro approach. Through scRNA-seq, differentially expressed genes between control and patient lines were identified. Some of these genes, linked to quantitative structural and functional changes, were cardiac specific, explaining the targeted nature of the disease progression seen in patients. The results of this work demonstrate the utility of combining in vitro tools in exploring heart disease mechanics.
Collapse
Affiliation(s)
- Mehrsa Mehrabi
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Tessa A Morris
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA.,Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA
| | - Zixuan Cang
- Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA.,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Cecilia H H Nguyen
- Genetics & Genomics Division, Department of Pediatrics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Yutong Sha
- Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Mira N Asad
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Nyree Khachikyan
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Taylor L Greene
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Danielle M Becker
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Qing Nie
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA.,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Michael V Zaragoza
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA.,Genetics & Genomics Division, Department of Pediatrics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Anna Grosberg
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA. .,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA. .,Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA. .,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA. .,Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA. .,The Henry Samueli School of Engineering, University of California, Irvine, 2418 Engineering Hall, Irvine, CA, 92697, USA.
| |
Collapse
|
13
|
Cai N, Gomez-Duran A, Yonova-Doing E, Kundu K, Burgess AI, Golder ZJ, Calabrese C, Bonder MJ, Camacho M, Lawson RA, Li L, Williams-Gray CH, Di Angelantonio E, Roberts DJ, Watkins NA, Ouwehand WH, Butterworth AS, Stewart ID, Pietzner M, Wareham NJ, Langenberg C, Danesh J, Walter K, Rothwell PM, Howson JMM, Stegle O, Chinnery PF, Soranzo N. Mitochondrial DNA variants modulate N-formylmethionine, proteostasis and risk of late-onset human diseases. Nat Med 2021; 27:1564-1575. [PMID: 34426706 DOI: 10.1038/s41591-021-01441-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 06/15/2021] [Indexed: 02/02/2023]
Abstract
Mitochondrial DNA (mtDNA) variants influence the risk of late-onset human diseases, but the reasons for this are poorly understood. Undertaking a hypothesis-free analysis of 5,689 blood-derived biomarkers with mtDNA variants in 16,220 healthy donors, here we show that variants defining mtDNA haplogroups Uk and H4 modulate the level of circulating N-formylmethionine (fMet), which initiates mitochondrial protein translation. In human cytoplasmic hybrid (cybrid) lines, fMet modulated both mitochondrial and cytosolic proteins on multiple levels, through transcription, post-translational modification and proteolysis by an N-degron pathway, abolishing known differences between mtDNA haplogroups. In a further 11,966 individuals, fMet levels contributed to all-cause mortality and the disease risk of several common cardiovascular disorders. Together, these findings indicate that fMet plays a key role in common age-related disease through pleiotropic effects on cell proteostasis.
Collapse
Affiliation(s)
- Na Cai
- Human Genetics Department, Wellcome Sanger Institute (WT), Hinxton, UK.,European Bioinformatics Institute (EMBL-EBI), Hinxton, UK.,Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Aurora Gomez-Duran
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CIB-CSIC), Madrid, Spain
| | - Ekaterina Yonova-Doing
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Primary Public Health and Primary Care, University of Cambridge, Cambridge, UK.,Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Kousik Kundu
- Human Genetics Department, Wellcome Sanger Institute (WT), Hinxton, UK
| | - Annette I Burgess
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Zoe J Golder
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Claudia Calabrese
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.,Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc J Bonder
- European Bioinformatics Institute (EMBL-EBI), Hinxton, UK.,Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marta Camacho
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Rachael A Lawson
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Lixin Li
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Caroline H Williams-Gray
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Primary Public Health and Primary Care, University of Cambridge, Cambridge, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK.,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - David J Roberts
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK.,NHS Blood and Transplant-Oxford Centre, John Radcliffe Hospital, Oxford, UK.,Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Nick A Watkins
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Willem H Ouwehand
- Human Genetics Department, Wellcome Sanger Institute (WT), Hinxton, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.,NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.,Department of Haematology, University of Cambridge, Cambridge, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Primary Public Health and Primary Care, University of Cambridge, Cambridge, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK.,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | | | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nick J Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - John Danesh
- Human Genetics Department, Wellcome Sanger Institute (WT), Hinxton, UK.,British Heart Foundation Cardiovascular Epidemiology Unit, Department of Primary Public Health and Primary Care, University of Cambridge, Cambridge, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK.,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK.,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Klaudia Walter
- Human Genetics Department, Wellcome Sanger Institute (WT), Hinxton, UK
| | - Peter M Rothwell
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Primary Public Health and Primary Care, University of Cambridge, Cambridge, UK.,Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Oliver Stegle
- European Bioinformatics Institute (EMBL-EBI), Hinxton, UK. .,Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK. .,Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| | - Nicole Soranzo
- Human Genetics Department, Wellcome Sanger Institute (WT), Hinxton, UK. .,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK. .,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK. .,Department of Haematology, University of Cambridge, Cambridge, UK. .,Genomics Research Centre, Human Technopole, Milan, Italy.
| |
Collapse
|
14
|
Friedrich VK, Rubel MA, Schurr TG. Mitochondrial genetic variation in human bioenergetics, adaptation, and adult disease. Am J Hum Biol 2021; 34:e23629. [PMID: 34146380 DOI: 10.1002/ajhb.23629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Mitochondria are critical for the survival of eukaryotic organisms due to their ability to produce cellular energy, which drives virtually all aspects of host biology. However, the effects of mitochondrial DNA (mtDNA) variation in relation to disease etiology and adaptation within contemporary global human populations remains incompletely understood. METHODS To develop a more holistic understanding of the role of mtDNA diversity in human adaptation, health, and disease, we investigated mitochondrial biology and bioenergetics. More specifically, we synthesized details from studies of mitochondrial function and variation in the context of haplogroup background, climatic adaptation, and oxidative disease. RESULTS The majority of studies show that mtDNA variation arose during modern human dispersal around the world. Some of these variants appear to have been positively selected for their adaptiveness in colder climates, with these sequence changes having implications for tissue-specific function and thermogenic capacity. In addition, many variants modulating energy production are also associated with damaging metabolic byproducts and mitochondrial dysfunction, which, in turn, are implicated in the onset and severity of several different adult mitochondrial diseases. Thus, mtDNA variation that governs bioenergetics, metabolism, and thermoregulation may potentially have adverse consequences for human health, depending on the genetic background and context in which it occurs. CONCLUSIONS Our review suggests that the mitochondrial research field would benefit from independently replicating mtDNA haplogroup-phenotype associations across global populations, incorporating potentially confounding environmental, demographic, and disease covariates into studies of mtDNA variation, and extending association-based studies to include analyses of complete mitogenomes and assays of mitochondrial function.
Collapse
Affiliation(s)
- Volney K Friedrich
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meagan A Rubel
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Translational Imaging and Precision Medicine, University of California - San Diego, La Jolla, California, USA
| | - Theodore G Schurr
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Kozakiewicz P, Grzybowska-Szatkowska L, Ciesielka M, Rzymowska J. The Role of Mitochondria in Carcinogenesis. Int J Mol Sci 2021; 22:ijms22105100. [PMID: 34065857 PMCID: PMC8151940 DOI: 10.3390/ijms22105100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
The mitochondria are essential for normal cell functioning. Changes in mitochondrial DNA (mtDNA) may affect the occurrence of some chronic diseases and cancer. This process is complex and not entirely understood. The assignment to a particular mitochondrial haplogroup may be a factor that either contributes to cancer development or reduces its likelihood. Mutations in mtDNA occurring via an increase in reactive oxygen species may favour the occurrence of further changes both in mitochondrial and nuclear DNA. Mitochondrial DNA mutations in postmitotic cells are not inherited, but may play a role both in initiation and progression of cancer. One of the first discovered polymorphisms associated with cancer was in the gene NADH-ubiquinone oxidoreductase chain 3 (mt-ND3) and it was typical of haplogroup N. In prostate cancer, these mutations and polymorphisms involve a gene encoding subunit I of respiratory complex IV cytochrome c oxidase subunit 1 gene (COI). At present, a growing number of studies also address the impact of mtDNA polymorphisms on prognosis in cancer patients. Some of the mitochondrial DNA polymorphisms occur in both chronic disease and cancer, for instance polymorphism G5913A characteristic of prostate cancer and hypertension.
Collapse
Affiliation(s)
- Paulina Kozakiewicz
- Department of Radiotherapy, Medical University in Lublin, Chodźki 7, 20-093 Lublin, Poland; (L.G.-S.); (M.C.)
- Department of Radiotherapy, St. John’s Cancer Centre, The Regional Oncology Centre of Lublin Jaczewskiego 7, 20-090 Lublin, Poland
- Correspondence:
| | - Ludmiła Grzybowska-Szatkowska
- Department of Radiotherapy, Medical University in Lublin, Chodźki 7, 20-093 Lublin, Poland; (L.G.-S.); (M.C.)
- Department of Radiotherapy, St. John’s Cancer Centre, The Regional Oncology Centre of Lublin Jaczewskiego 7, 20-090 Lublin, Poland
| | - Marzanna Ciesielka
- Department of Radiotherapy, Medical University in Lublin, Chodźki 7, 20-093 Lublin, Poland; (L.G.-S.); (M.C.)
- Chair and Department of Forensic Medicine, Medical University in Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Jolanta Rzymowska
- Chair and Department of Biology and Genetics, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland;
| |
Collapse
|
16
|
Chang X, Bakay M, Liu Y, Glessner J, Rathi KS, Hou C, Qu H, Vaksman Z, Nguyen K, Sleiman PMA, Diskin SJ, Maris JM, Hakonarson H. Mitochondrial DNA Haplogroups and Susceptibility to Neuroblastoma. J Natl Cancer Inst 2021; 112:1259-1266. [PMID: 32096864 DOI: 10.1093/jnci/djaa024] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/24/2020] [Accepted: 02/19/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neuroblastoma is a childhood malignancy that arises from the developing sympathetic nervous system. Although mitochondrial dysfunctions have been implicated in the pathophysiology of neuroblastoma, the role of mitochondrial DNA (mtDNA) has not been extensively investigated. METHODS A total of 2404 Caucasian children diagnosed with neuroblastoma and 9310 ancestry-matched controls were recruited at the Children's Hospital of Philadelphia. The mtDNA haplogroups were identified from SNP array data of two independent cohorts. We conducted a case-control study to explore potential associations of mtDNA haplogroups with the susceptibility of neuroblastoma. The genetic effect of neuroblastoma was measured by odds ratios (ORs) of mitochondrial haplogroups. All tests were two-sided. RESULTS Haplogroup K was statistically significantly associated with reduced risk of neuroblastoma in the discovery cohort consisting of 1474 cases and 5699 controls (OR = 0.72, 95% confidence interval [CI] = 0.57 to 0.90; P = 4.8 × 10-3). The association was replicated in an independent cohort (OR = 0.69, 95% CI = 0.53 to 0.92; P = .01) of 930 cases and 3611 controls. Pooled analysis was performed by combining the two data sets. The association remained highly statistically significant after correction for multiple testing (OR = 0.71, 95% CI = 0.59 to 0.84, P = 1.96 × 10-4, Pcorrected = .002). Further analysis focusing on neuroblastoma subtypes indicated haplogroup K was more associated with high-risk neuroblastoma (OR = 0.57, 95% CI = 0.43 to 0.76; P = 1.46 × 10-4) than low-risk and intermediate-risk neuroblastoma. CONCLUSIONS Haplogroup K is an independent genetic factor associated with reduced risk of developing neuroblastoma in European descents. These findings provide new insights into the genetic basis of neuroblastoma, implicating mitochondrial DNA encoded proteins in the etiology of neuroblastoma.
Collapse
Affiliation(s)
- Xiao Chang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marina Bakay
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yichuan Liu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joseph Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Komal S Rathi
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Cuiping Hou
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Huiqi Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zalman Vaksman
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kenny Nguyen
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Patrick M A Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sharon J Diskin
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.,Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
17
|
Association Study Between Genetic Variation in Whole Mitochondrial Genome and Ischemic Stroke. J Mol Neurosci 2021; 71:2152-2162. [PMID: 33447902 DOI: 10.1007/s12031-020-01778-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/11/2020] [Indexed: 10/22/2022]
Abstract
Mitochondrial DNA (mtDNA) affects the mitochondrial function, which is potentially related to susceptibility to ischemic stroke (IS). However, study on IS genetics by whole mitochondrial genome sequencing has not been extensively explored. Therefore, a two-stage study was designed to explore the relationship between the whole mitochondrial genome variants and IS. In the first stage, whole mitochondrial genomes of 52 IS patients and 55 controls were sequenced by next-generation sequencing. Fifty-three mtDNA mutation sites which may be related to the pathogenesis of IS were discovered. Nine unreported mtDNA variation sites were found for the first time. In the second larger Chinese cohort, we confirmed that m.T195C and m.T12338C in the mitochondrial D-loop region were the protective factors of IS, especially m.T195C and m.C311T in the LAA subtype. In conclusion, our study provided population genetic information and a reference for IS-relevant research, with wide applications in diagnosis, therapeutic treatments and prediction of IS.
Collapse
|
18
|
Gusdon AM, Hui Y, Chen J, Mathews CE, Qu S. Mitochondrial haplogroup G is associated with nonalcoholic fatty liver disease, while haplogroup A mitigates the effects of PNPLA3. Endocrinol Diabetes Metab 2021; 4:e00187. [PMID: 33532620 PMCID: PMC7831202 DOI: 10.1002/edm2.187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/05/2020] [Accepted: 08/29/2020] [Indexed: 12/17/2022] Open
Abstract
Objectives Mitochondrial dysfunction plays a pivotal role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). We hypothesized that mitochondrial DNA (mtDNA) haplogroups affect the risk of NAFLD in Han Chinese patients and interact with PNPLA3 genotypes. Design NAFLD and control patients were recruited from a tertiary care centre. The mitochondrial genome was amplified in overlapping segments and sequenced. Mitochondrial haplogroups were determined using Mitomaster. PNPLA3 rs738409 genotyping was performed using restriction fragment length polymorphism analysis. Patients We enrolled 655 NAFLD patients and 504 controls. Results More NAFLD patients encoded haplogroup G; odds ratio (OR) 1.85 (95% confidence interval [CI] 1.16, 2.80). Subhaplogroup G3 was present more frequently in NAFLD patients (25.8% vs 6.5%). The PNPLA3 CG genotype resulted in an OR of 1.66 (95% CI 1.25, 2.21), and the GG genotype resulted in an OR of 2.33 (95% CI 1.72, 3.17) for NAFLD. Patients with mitochondrial haplogroup A had a significantly higher frequency of genotype GG. Among patients with haplogroup A, no PNPLA3 genotype was associated with increased NAFLD risk (CG: OR 1.17, 95% CI 0.55, 2.34; GG: OR 1.04 95% CI 0.66, 2.65). Excluding haplogroup A, the OR for CG was 1.58 (95% CI 1.18, 2.12), and the OR for GG was 1.81 (95% CI 1.30, 2.51). Conclusion Haplogroup G was associated with an increased risk of NAFLD PNPLA3 GG genotype was overrepresented among patients encoding haplogroup A and was not associated with NAFLD risk among haplogroup A patients. Mitochondrial genetics influence NAFLD risk and interact with PNPLA3 genotypes.
Collapse
Affiliation(s)
- Aaron M. Gusdon
- Department of NeurosurgeryMischer Neuroscience AssociatesUniversity of Texas Health Science Center at HoustonHoustonTXUSA
| | - You Hui
- Department of EndocrinologyShanghai Tenth People's HospitalTongji UniversityShanghaiChina
| | - Jing Chen
- Department of Pathology, Immunology and Laboratory MedicineUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Clayton E. Mathews
- Department of Pathology, Immunology and Laboratory MedicineUniversity of Florida College of MedicineGainesvilleFLUSA
| | - Shen Qu
- Department of EndocrinologyShanghai Tenth People's HospitalTongji UniversityShanghaiChina
| |
Collapse
|
19
|
Mitochondrial DNA haplogroups and risk of attention deficit and hyperactivity disorder in European Americans. Transl Psychiatry 2020; 10:370. [PMID: 33139694 PMCID: PMC7608630 DOI: 10.1038/s41398-020-01064-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 09/13/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Although mitochondrial dysfunction has been implicated in the pathophysiology of attention deficit and hyperactivity disorder ADHD, the role of mitochondrial DNA (mtDNA) has not been extensively investigated. To determine whether mtDNA haplogroups influence risk of ADHD, we performed a case-control study comprising 2076 ADHD cases and 5078 healthy controls, all of whom were European decedents recruited from The Children's Hospital of Philadelphia (CHOP). Associations between eight major European mtDNA Haplogroups and ADHD risk were assessed in three independent European cohorts. Meta-analysis of the three studies indicated that mtDNA haplogroups K (odds ratio = 0.69, P = 2.24 × 10-4, Pcorrected = 1.79 × 10-3) and U (odds ratio = 0.77, P = 8.88 × 10-4, Pcorrected = 7.11 × 10-3) were significantly associated with reduced risk of ADHD. In contrast, haplogroup HHV* (odds ratio = 1.18, P = 2.32 × 10-3, Pcorrected = 0.019) was significantly associated with increased risk of ADHD. Our results provide novel insight into the genetic basis of ADHD, implicating mitochondrial mechanisms in the pathophysiology of this relatively common psychiatric disorder.
Collapse
|
20
|
Stewart JB, Chinnery PF. Extreme heterogeneity of human mitochondrial DNA from organelles to populations. Nat Rev Genet 2020; 22:106-118. [PMID: 32989265 DOI: 10.1038/s41576-020-00284-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
Contrary to the long-held view that most humans harbour only identical mitochondrial genomes, deep resequencing has uncovered unanticipated extreme genetic variation within mitochondrial DNA (mtDNA). Most, if not all, humans contain multiple mtDNA genotypes (heteroplasmy); specific patterns of variants accumulate in different tissues, including cancers, over time; and some variants are preferentially passed down or suppressed in the maternal germ line. These findings cast light on the origin and spread of mtDNA mutations at multiple scales, from the organelle to the human population, and challenge the conventional view that high percentages of a mutation are required before a new variant has functional consequences.
Collapse
Affiliation(s)
- James B Stewart
- Max Planck Institute for Biology of Ageing, Cologne, Germany.,Wellcome Centre for Mitochondrial Research, Newcastle University Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
21
|
Ischemic Stroke Risk Associated with Mitochondrial Haplogroup F in the Asian Population. Cells 2020; 9:cells9081885. [PMID: 32796743 PMCID: PMC7463505 DOI: 10.3390/cells9081885] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
Mitochondrial dysfunction is involved in the pathogenesis of atherosclerosis, the primary risk factor for ischemic stroke. This study aims to explore the role of mitochondrial genomic variations in ischemic stroke, and to uncover the nuclear genes involved in this relationship. Eight hundred and thirty Taiwanese patients with a history of ischemic stroke and 966 normal controls were genotyped for their mitochondrial haplogroup (Mthapg). Cytoplasmic hybrid cells (cybrids) harboring different Mthapgs were used to observe functional differences under hypoxia-ischemia. RNA sequencing (RNASeq) was conducted to identify the particularly elevated mRNA. The patient study identified an association between Mthapg F1 and risk of ischemic stroke (OR 1.72:1.27-2.34, p = 0.001). The cellular study further demonstrated an impeded induction of hypoxic inducible factor 1α in the Mthapg F1 cybrid after hypoxia-ischemia. Additionally, the study demonstrated that Mthapg F cybrids were associated with an altered mitochondrial function, including decreased oxygen consumption, higher mitochondrial ROS production, and lower mitochondrial membrane potential. Mthapg F cybrids were also noted to be prone to inflammation, with increased expression of several inflammatory cytokines and elevated matrix metalloproteinase 9. The RNASeq identified significantly elevated expressions of angiopoietin-like 4 in Mthapg F1 cybrids after hypoxia-ischemia. Our study demonstrates an association between Mthapg F and susceptibility to ischemic stroke.
Collapse
|
22
|
Hagen CM, Elson JL, Hedley PL, Aidt FH, Havndrup O, Jensen MK, Kanters JK, Atherton JJ, McGaughran J, Bundgaard H, Christiansen M. Evolutionary dissection of mtDNA hg H: a susceptibility factor for hypertrophic cardiomyopathy. Mitochondrial DNA A DNA Mapp Seq Anal 2020; 31:238-244. [PMID: 32602800 DOI: 10.1080/24701394.2020.1782897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mitochondrial DNA (mtDNA) haplogroup (hg) H has been reported as a susceptibility factor for hypertrophic cardiomyopathy (HCM). This was established in genetic association studies, however, the SNP or SNP's that are associated with the increased risk have not been identified. Hg H is the most frequent European mtDNA hg with greater than 80 subhaplogroups (subhgs) each defined by specific SNPs. We tested the hypothesis that the distribution of H subhgs might differ between HCM patients and controls. The subhg H distribution in 55 HCM index cases was compared to that of two Danish mtDNA hg H control groups (n = 170 and n = 908, respectively). In the HCM group, H and 12 different H subhgs were found. All these, except subhgs H73, were also found in both control groups. The HCM group was also characterized by a higher proportion of H3 compared to H2. In the HCM group the H3/H2 proportion was 1.7, whereas it was 0.45 and 0.54 in the control groups. This tendency was replicated in an independent group of Hg H HCM index cases (n = 39) from Queensland, Australia, where the H3/H2 ratio was 1.5. In conclusion, the H subhgs distribution differs between HCM cases and controls, but the difference is subtle, and the understanding of the pathogenic significance is hampered by the lack of functional studies on the subhgs of H.
Collapse
Affiliation(s)
- Christian M Hagen
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joanna L Elson
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark.,Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Paula L Hedley
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
| | - Frederik H Aidt
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
| | - Ole Havndrup
- Department of Cardiology, Roskilde Hospital, Roskilde, Denmark
| | - Morten K Jensen
- Department of Medicine B, The Heart Center, Copenhagen, Denmark
| | - Jørgen K Kanters
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - John J Atherton
- Department of Cardiology, Royal Brisbane Hospital and School of Medicine, University of Queensland, Brisbane, Australia
| | - Julie McGaughran
- Queensland Clinical Genetics Service, Royal Children's Hospital and School of Medicine, Brisbane, Australia
| | | | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
The alterations of mitochondrial DNA in coronary heart disease. Exp Mol Pathol 2020; 114:104412. [PMID: 32113905 DOI: 10.1016/j.yexmp.2020.104412] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 02/24/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022]
Abstract
Coronary heart disease (CHD) is the major cause of death in modern society. CHD is characterized by atherosclerosis, which could lead to vascular cavity stenosis or obstruction, resulting in ischemic cardiac conditions such as angina and myocardial infarction. In terms of the mitochondrion, the main function is to produce adenosine triphosphate (ATP) for cells. And the alterations (including mutations, altered copy number and haplogroups) of mitochondrial DNA (mtDNA) are associated with the abnormal expression of oxidative phosphorylation (OXPHOS) system, resulting in mitochondrial dysfunction, then leading to perturbation on the electron transport chain and increased ROS generation and reduction in ATP level, contributing to ATP-producing disorders and oxidative stress, which may further accelerate development or vulnerability of atherosclerosis and myocardial ischemic injury. Therefore, the mtDNA defects may play an important role in making an early diagnosis, identifying disease-specific biomarkers and therapeutic targets, and predicting outcomes for patients with atherosclerosis and CHD. In this review, we aim to summarize the contribution of mtDNA mutations, altered mtDNA copy number and mtDNA haplogroups on the occurrence and development of CHD.
Collapse
|
24
|
Veronese N, Stubbs B, Koyanagi A, Vaona A, Demurtas J, Schofield P, Maggi S. Mitochondrial genetic haplogroups and cardiovascular diseases: Data from the Osteoarthritis Initiative. PLoS One 2019; 14:e0213656. [PMID: 30921349 PMCID: PMC6438497 DOI: 10.1371/journal.pone.0213656] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 02/26/2019] [Indexed: 12/13/2022] Open
Abstract
Background Some case-control studies reported that mitochondrial haplogroups could be associated with the onset of cardiovascular diseases (CVD), but the literature regarding this topic is limited. We aimed to investigate whether any mitochondrial haplogroup carried a higher or lower risk of CVD in a large cohort of North American people affected by knee osteoarthritis or at high risk for this condition. Materials and methods A longitudinal cohort study including individuals from the Osteoarthritis Initiative was done. Haplogroups were assigned through a combination of sequencing and PCR-RFLP techniques. All the mitochondrial haplogroups have been named following this nomenclature: HV, JT, UK, IWX, and superHV/others. The strength of the association between mitochondrial haplogroups and incident CVD was evaluated through a Cox’s regression analysis, adjusted for potential confounders, and reported as hazard ratios (HRs) with their 95% confidence intervals (CIs). Results Overall, 3,288 Caucasian participants (56.8% women) with a mean age of 61.3±9.2 years without CVD at baseline were included. During a median follow-up of 8 years, 322 individuals (= 9.8% of baseline population) developed a CVD. After adjusting for 11 potential confounders at baseline and taking those with the HV haplotype as reference (the most frequent), those with JT carried a significant lower risk of CVD (HR = 0.75; 95%CI: 0.54–0.96; p = 0.03). Participants with the J haplogroup had the lowest risk of CVD (HR = 0.71; 95%CI: 0.46–0.95; p = 0.02). Conclusions The presence of JT haplogroups (particularly J) may be associated with a reduced risk of CVD. However, this result was not based on a high level of statistical significance. Thus, future research with larger sample size is needed to assess whether our results can be corroborated.
Collapse
Affiliation(s)
- Nicola Veronese
- National Research Council, Neuroscience Institute, Aging Branch, Padova, National Institute of Gastroenterology “S. De Bellis” Research Hospital, Castellana Grotte (Ba), Italy
- * E-mail:
| | - Brendon Stubbs
- South London and Maudsley NHS Foundation Trust, Denmark Hill, London, United Kingdom
| | - Ai Koyanagi
- Research and Development Unit, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, CIBERSAM, Sant Boi de Llobregat, Barcelona, Spain
| | - Alberto Vaona
- Primary Care Department, Azienda ULSS20 Verona, Verona, Italy
| | - Jacopo Demurtas
- Primary Care Department, Azienda USL Toscana Sud Est, Grosseto, Italy
| | - Patricia Schofield
- Faculty of Health, Social Care and Education, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Stefania Maggi
- National Research Council, Neuroscience Institute, Aging Branch, Padova, National Institute of Gastroenterology “S. De Bellis” Research Hospital, Castellana Grotte (Ba), Italy
| |
Collapse
|
25
|
Venter M, Tomas C, Pienaar IS, Strassheim V, Erasmus E, Ng WF, Howell N, Newton JL, Van der Westhuizen FH, Elson JL. MtDNA population variation in Myalgic encephalomyelitis/Chronic fatigue syndrome in two populations: a study of mildly deleterious variants. Sci Rep 2019; 9:2914. [PMID: 30814539 PMCID: PMC6393470 DOI: 10.1038/s41598-019-39060-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/11/2019] [Indexed: 02/07/2023] Open
Abstract
Myalgic Encephalomyelitis (ME), also known as Chronic Fatigue Syndrome (CFS) is a debilitating condition. There is growing interest in a possible etiologic or pathogenic role of mitochondrial dysfunction and mitochondrial DNA (mtDNA) variation in ME/CFS. Supporting such a link, fatigue is common and often severe in patients with mitochondrial disease. We investigate the role of mtDNA variation in ME/CFS. No proven pathogenic mtDNA mutations were found. We then investigated population variation. Two cohorts were analysed, one from the UK (n = 89 moderately affected; 29 severely affected) and the other from South Africa (n = 143 moderately affected). For both cohorts, ME/CFS patients had an excess of individuals without a mildly deleterious population variant. The differences in population variation might reflect a mechanism important to the pathophysiology of ME/CFS.
Collapse
Affiliation(s)
- Marianne Venter
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Cara Tomas
- Institute of Cellular Medicine & NIHR Biomedical Research Centre in Ageing and Chronic Disease, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Ilse S Pienaar
- School of Life Sciences, University of Sussex, Falmer, BN1 9PH, United Kingdom
- Centre for Neuroinflammation and Neurodegeneration, Imperial College London, London, United Kingdom
| | - Victoria Strassheim
- Institute of Cellular Medicine & NIHR Biomedical Research Centre in Ageing and Chronic Disease, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Centre for Neuroinflammation and Neurodegeneration, Imperial College London, London, United Kingdom
| | - Elardus Erasmus
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Wan-Fai Ng
- Institute of Cellular Medicine & NIHR Biomedical Research Centre in Ageing and Chronic Disease, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Neil Howell
- Department of Radiation Therapy, UTMB, Galveston, Texas, USA
| | - Julia L Newton
- Centre for Neuroinflammation and Neurodegeneration, Imperial College London, London, United Kingdom
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | | | - Joanna L Elson
- Human Metabolomics, North-West University, Potchefstroom, South Africa.
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom.
| |
Collapse
|
26
|
Mitochondrial Haplogroup and the Risk of Acute Kidney Injury Following Cardiac Bypass Surgery. Sci Rep 2019; 9:2279. [PMID: 30783114 PMCID: PMC6381211 DOI: 10.1038/s41598-018-37944-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 12/11/2018] [Indexed: 02/02/2023] Open
Abstract
Although mitochondrial dysfunction plays a key role in the pathophysiology of acute kidney injury (AKI), the influence of mitochondrial genetic variability in this process remains unclear. We explored the association between the risk of post-cardiac bypass AKI and mitochondrial haplotype - inherited mitochondrial genomic variations of potentially functional significance. Our single-centre study recruited consecutive patients prior to surgery. Exclusions included stage 5 CKD, non-Caucasian race and subsequent off-pump surgery. Haplogroup analysis allowed characterisation of the study population using the common mutations and by phylogenetic supergroup (WXI and HV). Chi-square tests for association allowed the identification of potential predictors of AKI for use in logistic regression analysis. AKI occurred in 12.8% of the study population (n = 881; male 69.6%, non-diabetic 78.5%, median (interquartile range) age 68.0 (61.0-75.0) years). The haplogroup profile comprised H (42.7%), J (12.1%), T (10.9%), U (14.4%) and K (7.6%). Although the regression model was statistically significant (χ2 = 95.483, p < 0.0005), neither the phylogenetic supergroups nor any individual haplogroup was a significant contributor. We found no significant association between common European haplogroups and the risk of post-cardiac bypass AKI. However, given the major role of mitochondrial dysfunction in AKI, there is a need to replicate our findings in other cohorts and with other aetiologies of AKI.
Collapse
|
27
|
Chalkia D, Chang YC, Derbeneva O, Lvova M, Wang P, Mishmar D, Liu X, Singh LN, Chuang LM, Wallace DC. Mitochondrial DNA associations with East Asian metabolic syndrome. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2018; 1859:878-892. [PMID: 29997041 PMCID: PMC6530988 DOI: 10.1016/j.bbabio.2018.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/04/2018] [Accepted: 07/04/2018] [Indexed: 01/31/2023]
Abstract
Mitochondrial dysfunction has repeatedly been reported associated with type 2 diabetes mellitus (T2DM) and metabolic syndrome (MS), as have mitochondrial DNA (mtDNA) tRNA and duplication mutations and mtDNA haplogroup lineages. We identified 19 Taiwanese T2DM and MS pedigrees from Taiwan, with putative matrilineal transmission, one of which harbored the pathogenic mtDNA tRNALeu(UUR) nucleotide (nt) 3243A>G mutation on the N9a3 haplogroup background. We then recruited three independent Taiwanese cohorts, two from Taipei (N = 498, mean age 52 and N = 1002, mean age 44) and one from a non-urban environment (N = 501, mean age 57). All three cohorts were assessed for an array of metabolic parameters, their mtDNA haplogroups determined, and the haplogroups correlated with T2DM/MS phenotypes. Logistic regression analysis revealed that mtDNA haplogroups D5, F4, and N9a conferred T2DM protection, while haplogroups F4 and N9a were risk factors for hypertension (HTN), and F4 was a risk factor for obesity (OB). Additionally, the 5263C>T (ND2 A165V) variant commonly associated with F4 was associated with hypertension (HTN). Cybrids were prepared with macro-haplogroup N (defined by variants m.ND3 10398A (114T) and m.ATP6 8701A (59T)) haplogroups B4 and F1 mtDNAs and from macro-haplogroup M (variants m.ND3 10398G (114A) and m.ATP6 8701G (59A)) haplogroup M9 mtDNAs. Additionally, haplogroup B4 and F1 cybrids were prepared with and without the mtDNA variant in ND1 3394T>C (Y30H) reported to be associated with T2DM. Assay of mitochondria complex I in these cybrids revealed that macro-haplogroup N cybrids had lower activity than M cybrids, that haplogroup F cybrids had lower activity than B4 cybrids, and that the ND1 3394T>C (Y30H) variant reduced complex I on both the B4 and F1 background but with very different cumulative effects. These data support the hypothesis that functional mtDNA variants may contribute to the risk of developing T2DM and MS.
Collapse
Affiliation(s)
- Dimitra Chalkia
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America
| | - Yi-Cheng Chang
- Department of Internal Medicine, National Taiwan University Medical College, Taipei, Taiwan; Graduate Institute of Medical Genomics and Proteomics, National Taiwan University Medical College, Taipei, Taiwan; Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Olga Derbeneva
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Maria Lvova
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America
| | - Ping Wang
- Department of Medicine, University of California, Irvine School of Medicine, Irvine, CA 92697, United States of America
| | - Dan Mishmar
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Xiaogang Liu
- Douglas C. Wallace Institute for Mitochondrial and Epigenomic Information Sciences, The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China
| | - Larry N Singh
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Medical College, Taipei, Taiwan
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States of America; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America; Douglas C. Wallace Institute for Mitochondrial and Epigenomic Information Sciences, The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; Institute of Molecular Genetics, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
28
|
Riley V, Erzurumluoglu AM, Rodriguez S, Bonilla C. Mitochondrial DNA Haplogroups and Breast Cancer Risk Factors in the Avon Longitudinal Study of Parents and Children (ALSPAC). Genes (Basel) 2018; 9:E395. [PMID: 30071701 PMCID: PMC6115984 DOI: 10.3390/genes9080395] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 01/11/2023] Open
Abstract
The relationship between mitochondrial DNA (mtDNA) and breast cancer has been frequently examined, particularly in European populations. However, studies reporting associations between mtDNA haplogroups and breast cancer risk have had a few shortcomings including small sample sizes, failure to account for population stratification and performing inadequate statistical tests. In this study we investigated the association of mtDNA haplogroups of European origin with several breast cancer risk factors in mothers and children of the Avon Longitudinal Study of Parents and Children (ALSPAC), a birth cohort that enrolled over 14,000 pregnant women in the Southwest region of the UK. Risk factor data were obtained from questionnaires, clinic visits and blood measurements. Information on over 40 independent breast cancer risk factor-related variables was available for up to 7781 mothers and children with mtDNA haplogroup data in ALSPAC. Linear and logistic regression models adjusted for age, sex and population stratification principal components were evaluated. After correction for multiple testing we found no evidence of association of European mtDNA haplogroups with any of the breast cancer risk factors analysed. Mitochondrial DNA haplogroups are unlikely to underlie susceptibility to breast cancer that occurs via the risk factors examined in this study of a population of European ancestry.
Collapse
Affiliation(s)
- Vivienne Riley
- MSc Genomic Medicine Programme, G7, College House, St Luke's Campus University of Exeter, Exeter, Devon EX2 4TE, UK.
| | - A Mesut Erzurumluoglu
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK.
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.
| | - Santiago Rodriguez
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.
| | - Carolina Bonilla
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.
- Integrative Cancer Epidemiology Program, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.
- Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil.
| |
Collapse
|
29
|
Ji F, Zhao C, Wang B, Tang Y, Miao Z, Wang Y. The role of 5-hydroxymethylcytosine in mitochondria after ischemic stroke. J Neurosci Res 2018; 96:1717-1726. [PMID: 30043506 DOI: 10.1002/jnr.24274] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 12/29/2022]
Abstract
5-Hydroxymethylcytosine (5hmC) exists in DNA, RNA, and mitochondrial DNA (mtDNA) and plays an important role in many diseases. Specifically, 5hmC is involved in promoting gene expression, and this process is regulated by Tet enzymes. In this study, we identified that there is no difference in male mice and female mice at first; then we examined the levels of 5hmC in mtDNA and explored the relationship among 5hmC, mitochondrial gene expression and ATP production after acute brain ischemia. The abundance of mtDNA 5hmC was increased at 1 d and peaked at 2 d after ischemic injury, whereas that of mtDNA 5mC was unchanged. Furthermore, increased mitochondrial Tet2 expression was found to be responsible for the increase in mtDNA 5hmC. Tet2 inhibition decreased the mtDNA 5hmC abundance and increased the ATP levels in mitochondria, suggesting an association between the cellular ATP levels and mtDNA 5hmC abundance. We also demonstrated that mtDNA 5hmC increased the mRNA levels of mitochondrial genes after ischemia/reperfusion (I/R) injury.
Collapse
Affiliation(s)
- Feng Ji
- Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Chenyu Zhao
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou City, China
| | - Bin Wang
- Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Yan Tang
- Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Yongxiang Wang
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou City, China.,Department of Orthopedics, Northern Jiangsu People's Hospital, Yangzhou City, China
| |
Collapse
|
30
|
Alawieh A, Zhao J, Feng W. Factors affecting post-stroke motor recovery: Implications on neurotherapy after brain injury. Behav Brain Res 2018; 340:94-101. [PMID: 27531500 PMCID: PMC5305670 DOI: 10.1016/j.bbr.2016.08.029] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/27/2016] [Accepted: 08/12/2016] [Indexed: 02/05/2023]
Abstract
Neurological disorders are a major cause of chronic disability globally among which stroke is a leading cause of chronic disability. The advances in the medical management of stroke patients over the past decade have significantly reduced mortality, but at the same time increased numbers of disabled survivors. Unfortunately, this reduction in mortality was not paralleled by satisfactory therapeutics and rehabilitation strategies that can improve functional recovery of patients. Motor recovery after brain injury is a complex, dynamic, and multifactorial process in which an interplay among genetic, pathophysiologic, sociodemographic and therapeutic factors determines the overall recovery trajectory. Although stroke recovery is the most well-studied form of post-injury neuronal recovery, a thorough understanding of the pathophysiology and determinants affecting stroke recovery is still lacking. Understanding the different variables affecting brain recovery after stroke will not only provide an opportunity to develop therapeutic interventions but also allow for developing personalized platforms for patient stratification and prognosis. We aim to provide a narrative review of major determinants for post-stroke recovery and their implications in other forms of brain injury.
Collapse
Affiliation(s)
- Ali Alawieh
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jing Zhao
- Minhang District Central Hospital, Fudan University, Shanghai, 201199, China
| | - Wuwei Feng
- Department of Neurology, MUSC Stroke Center, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Health Science and Research, The Center of Rehabilitation Science in Neurological Conditions, College of Health Professions, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
31
|
Guyatt AL, Burrows K, Guthrie PAI, Ring S, McArdle W, Day INM, Ascione R, Lawlor DA, Gaunt TR, Rodriguez S. Cardiometabolic phenotypes and mitochondrial DNA copy number in two cohorts of UK women. Mitochondrion 2018; 39:9-19. [PMID: 28818596 PMCID: PMC5832987 DOI: 10.1016/j.mito.2017.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/06/2017] [Accepted: 08/10/2017] [Indexed: 12/14/2022]
Abstract
The mitochondrial genome is present at variable copy number between individuals. Mitochondria are vulnerable to oxidative stress, and their dysfunction may be associated with cardiovascular disease. The association of mitochondrial DNA copy number with cardiometabolic risk factors (lipids, glycaemic traits, inflammatory markers, anthropometry and blood pressure) was assessed in two independent cohorts of European origin women, one in whom outcomes were measured at mean (SD) age 30 (4.3) years (N=2278) and the second at 69.4 (5.5) years (N=2872). Mitochondrial DNA copy number was assayed by quantitative polymerase chain reaction. Associations were adjusted for smoking, sociodemographic status, laboratory factors and white cell traits. Out of a total of 12 outcomes assessed in both cohorts, mitochondrial DNA copy number showed little or no association with the majority (point estimates were close to zero and nearly all p-values were >0.01). The strongest evidence was for an inverse association in the older cohort with insulin (standardised beta [95%CI]: -0.06, [-0.098, -0.022], p=0.002), but this association did not replicate in the younger cohort. Our findings do not provide support for variation in mitochondrial DNA copy number having an important impact on cardio-metabolic risk factors in European origin women.
Collapse
Affiliation(s)
- Anna L Guyatt
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Kimberley Burrows
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Philip A I Guthrie
- School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Sue Ring
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Wendy McArdle
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Ian N M Day
- School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Raimondo Ascione
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Santiago Rodriguez
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.
| |
Collapse
|
32
|
Umbria M, Ramos A, Caner J, Vega T, Lozano JE, Santos C, Aluja MP. Involvement of mitochondrial haplogroups in myocardial infarction and stroke: A case-control study in Castile and Leon (Spain) population. Mitochondrion 2017; 44:1-6. [PMID: 29258787 DOI: 10.1016/j.mito.2017.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 10/05/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022]
Abstract
There are strong evidences that common mitochondrial DNA (mtDNA) haplogroups may influence the pathogenesis of cardiovascular diseases (CVDs). In this matched case-control study, we investigate the association between mtDNA haplogroups and two CVDs, myocardial infarction (MI) and stroke, and classical cardiovascular risk factors. Data obtained show that haplogroup H constitute a susceptibility risk factor for MI (p=0.001; OR=2.379, 95% CI [1.440-3.990]). Otherwise, our data also suggest a beneficial role of haplogroup J against hypertension (p=0.019; OR=0.348, 95% CI [0.144-0.840]). These results may provide some guidance for predicting the genetic risk of these diseases in different human populations through the differences in energy efficiency between haplogroups.
Collapse
Affiliation(s)
- Miriam Umbria
- Unitat d'Antropologia Biològica, Department BABVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Amanda Ramos
- Unitat d'Antropologia Biològica, Department BABVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Jennifer Caner
- Unitat d'Antropologia Biològica, Department BABVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Tomás Vega
- Dirección General de Salud Pública, Consejería de Sanidad, Junta de Castilla y León, 47007 Valladolid, Spain
| | - José Eugenio Lozano
- Dirección General de Salud Pública, Consejería de Sanidad, Junta de Castilla y León, 47007 Valladolid, Spain
| | - Cristina Santos
- Unitat d'Antropologia Biològica, Department BABVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Maria Pilar Aluja
- Unitat d'Antropologia Biològica, Department BABVE, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
33
|
Lien LM, Chiou HY, Yeh HL, Chiu SY, Jeng JS, Lin HJ, Hu CJ, Hsieh FI, Wei YH. Significant Association Between Low Mitochondrial DNA Content in Peripheral Blood Leukocytes and Ischemic Stroke. J Am Heart Assoc 2017; 6:JAHA.117.006157. [PMID: 29151031 PMCID: PMC5721740 DOI: 10.1161/jaha.117.006157] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background Cumulative evidence has shown that low mitochondrial DNA (mtDNA) content is related to elevated oxidative stress and atherosclerosis, which play important roles in ischemic stroke. The objective of this study was to explore the association between mtDNA content in peripheral blood leukocytes and ischemic stroke. Methods and Results A total of 350 patients with first‐ever ischemic stroke and 350 healthy controls were recruited in this case‐control study. The mtDNA content in peripheral blood leukocytes was determined by quantitative real‐time polymerase chain reaction. The levels of oxidized glutathione, reduced glutathione, and 8‐hydroxy‐2′‐deoxyguanosine were measured by ELISA kits. Multivariate logistic regression models were used to analyze the relationship between mtDNA content in peripheral blood leukocytes and ischemic stroke. Our results show that mtDNA content of patients with ischemic stroke was notably lower compared with controls. A significant association was found between low mtDNA content and ischemic stroke. Furthermore, significant interactions were identified between low mtDNA and proven risk factors in patients with ischemic stroke. The levels of oxidized glutathione and 8‐hydroxy‐2′‐deoxyguanosine were significantly greater in patients with ischemic stroke compared with controls. Conclusions Our results demonstrate that low mtDNA content in peripheral blood leukocytes is associated with ischemic stroke. The relationship of low mtDNA content and ischemic stroke was particularly notable in individuals who had low mtDNA content combined with diabetes mellitus, metabolic syndrome, or cigarette smoking. Oxidative stress may be one of the contributory factors to decreased mtDNA content in patients with ischemic stroke.
Collapse
Affiliation(s)
- Li-Ming Lien
- Department of Neurology, Shin-Kong WHS Memorial Hospital, Taipei, Taiwan.,School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yi Chiou
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Hsu-Ling Yeh
- Department of Neurology, Shin-Kong WHS Memorial Hospital, Taipei, Taiwan.,School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shang-Yen Chiu
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Stroke Center and Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Huey-Juan Lin
- Department of Neurology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chaur-Jong Hu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Fang-I Hsieh
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Yau-Huei Wei
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City, Taiwan
| |
Collapse
|
34
|
Venter M, van der Westhuizen FH, Elson JL. The aetiology of cardiovascular disease: a role for mitochondrial DNA? Cardiovasc J Afr 2017; 29:122-132. [PMID: 28906532 PMCID: PMC6009096 DOI: 10.5830/cvja-2017-037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 07/17/2017] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular disease (CVD) is a world-wide cause of mortality in humans and its incidence is on the rise in Africa. In this review, we discuss the putative role of mitochondrial dysfunction in the aetiology of CVD and consequently identify mitochondrial DNA (mtDNA) variation as a viable genetic risk factor to be considered. We then describe the contribution and pitfalls of several current approaches used when investigating mtDNA in relation to complex disease. We also propose an alternative approach, the adjusted mutational load hypothesis, which would have greater statistical power with cohorts of moderate size, and is less likely to be affected by population stratification. We therefore address some of the shortcomings of the current haplogroup association approach. Finally, we discuss the unique challenges faced by studies done on African populations, and recommend the most viable methods to use when investigating mtDNA variation in CVD and other common complex disease.
Collapse
Affiliation(s)
- Marianne Venter
- Human Metabolomics, North-West University, Potchefstroom, South Africa.
| | | | - Joanna L Elson
- Human Metabolomics, North-West University, Potchefstroom, South Africa; Institute of Genetic Medicine, Newcastle University, United Kingdom
| |
Collapse
|
35
|
Gender, aging and longevity in humans: an update of an intriguing/neglected scenario paving the way to a gender-specific medicine. Clin Sci (Lond) 2017; 130:1711-25. [PMID: 27555614 PMCID: PMC4994139 DOI: 10.1042/cs20160004] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 06/27/2016] [Indexed: 12/25/2022]
Abstract
Data showing a remarkable gender difference in life expectancy and mortality, including survival to extreme age, are reviewed starting from clinical and demographic data and stressing the importance of a comprehensive historical perspective and a gene–environment/lifestyle interaction. Gender difference regarding prevalence and incidence of the most important age-related diseases, such as cardiovascular and neurodegenerative diseases, cancer, Type 2 diabetes, disability, autoimmunity and infections, are reviewed and updated with particular attention to the role of the immune system and immunosenescence. On the whole, gender differences appear to be pervasive and still poorly considered and investigated despite their biomedical relevance. The basic biological mechanisms responsible for gender differences in aging and longevity are quite complex and still poorly understood. The present review focuses on centenarians and their offspring as a model of healthy aging and summarizes available knowledge on three basic biological phenomena, i.e. age-related X chromosome inactivation skewing, gut microbiome changes and maternally inherited mitochondrial DNA genetic variants. In conclusion, an appropriate gender-specific medicine approach is urgently needed and should be systematically pursued in studies on healthy aging, longevity and age-related diseases, in a globalized world characterized by great gender differences which have a high impact on health and diseases.
Collapse
|
36
|
Abstract
Ischaemic heart disease and stroke are vascular events with serious health consequences worldwide. Recent genetic and epigenetic techniques have revealed many genetic determinants of these vascular events and simplified the approaches to research focused on ischaemic heart disease and stroke. The pathogenetic mechanisms of ischaemic heart disease and stroke are complex, with mitochondrial involvement (partially or entirely) recently gaining substantial support. Not only can mitochondrial reactive oxygen species give rise to ischaemic heart disease and stroke by production of oxidised low-density lipoprotein and induction of apoptosis, but the impact on pericytes contributes directly to the pathogenesis. Over the past two decades, publications implicate the causative role of nuclear genes in the development of ischaemic heart disease and stroke, in contrast to the potential role of mitochondrial DNA (mtDNA) in the pathophysiology of the disorders, which is much less understood, although recent studies do demonstrate that the involvement of mitochondria and mtDNA in the development of ischaemic heart disease and stroke is likely to be larger than originally thought, with the novel discovery of links among mitochondria, mtDNA and vascular events. Here we explore the molecular events and mtDNA alterations in relation to the role of mitochondria in ischaemic heart disease and stroke.
Collapse
|
37
|
Venter M, Malan L, van Dyk E, Elson JL, van der Westhuizen FH. Using MutPred derived mtDNA load scores to evaluate mtDNA variation in hypertension and diabetes in a two-population cohort: The SABPA study. J Genet Genomics 2016; 44:139-149. [PMID: 28298255 DOI: 10.1016/j.jgg.2016.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/11/2016] [Accepted: 12/22/2016] [Indexed: 11/20/2022]
Abstract
Mitochondrial DNA (mtDNA) variation has been implicated in many common complex diseases, but inconsistent and contradicting results are common. Here we introduce a novel mutational load hypothesis, which also considers the collective effect of mainly rare variants, utilising the MutPred Program. We apply this new methodology to investigate the possible role of mtDNA in two cardiovascular disease (CVD) phenotypes (hypertension and hyperglycaemia), within a two-population cohort (n = 363; mean age 45 ± 9 yrs). Very few studies have looked at African mtDNA variation in the context of complex disease, and none using complete sequence data in a well-phenotyped cohort. As such, our study will also extend our knowledge of African mtDNA variation, with complete sequences of Southern Africans being especially under-represented. The cohort showed prevalence rates for hypertension (58.6%) and prediabetes (44.8%). We could not identify a statistically significant role for mtDNA variation in association with hypertension or hyperglycaemia in our cohort. However, we are of the opinion that the method described will find wide application in the field, being especially useful for cohorts from multiple locations or with a variety of mtDNA lineages, where the traditional haplogroup association method has been particularly likely to generate spurious results in the context of association with common complex disease.
Collapse
Affiliation(s)
- Marianne Venter
- Human Metabolomics, North-West University, Potchefstroom 2531, South Africa
| | - Leone Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom 2531, South Africa
| | - Etresia van Dyk
- Human Metabolomics, North-West University, Potchefstroom 2531, South Africa
| | - Joanna L Elson
- Human Metabolomics, North-West University, Potchefstroom 2531, South Africa; Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom.
| | | |
Collapse
|
38
|
Mitochondrial genome association study with peripheral arterial disease and venous thromboembolism. Atherosclerosis 2016; 252:97-105. [DOI: 10.1016/j.atherosclerosis.2016.07.920] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/06/2016] [Accepted: 07/26/2016] [Indexed: 11/17/2022]
|
39
|
Siegismund CS, Schäfer I, Seibel P, Kühl U, Schultheiss HP, Lassner D. Mitochondrial haplogroups and expression studies of commonly used human cell lines. Mitochondrion 2016; 30:236-47. [PMID: 27562426 DOI: 10.1016/j.mito.2016.08.012] [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: 03/01/2016] [Revised: 07/29/2016] [Accepted: 08/19/2016] [Indexed: 02/02/2023]
Abstract
We developed a multiplex fragment length analysis (MFLA) for clearly assigning mitochondrial haplogroups mostly endemic in Europe for future cardiac diagnostics. As a technical proof, 23 commonly used human cell lines were haplotyped as reference standards. The functional analysis on mtDNA copies per cell revealed no correlation to haplogroups but a relatively high rate of mitochondria per cell and at the same time a very low expression of all mitochondrial and some nuclear encoded mitochondrial related genes. Established MFLA is an easy to handle method for analysing European mitochondrial haplogroups to perform epidemic studies and elucidate correlations to distinct diseases.
Collapse
Affiliation(s)
| | - Ingo Schäfer
- University of Leipzig, Centre for Biotechnology and Biomedicine (BBZ), Molecular Cell Therapy, Deutscher Platz 5, D-04103 Leipzig, Germany
| | - Peter Seibel
- University of Leipzig, Centre for Biotechnology and Biomedicine (BBZ), Molecular Cell Therapy, Deutscher Platz 5, D-04103 Leipzig, Germany
| | - Uwe Kühl
- Institute for Cardiac Diagnostics and Therapy (IKDT), Moltkestr. 31, D-12203 Berlin, Germany; Department of Cardiology, Campus Virchow, Charité - University Hospital Berlin, Augustenburger Platz 1, D-13353, Germany
| | - Heinz-Peter Schultheiss
- Institute for Cardiac Diagnostics and Therapy (IKDT), Moltkestr. 31, D-12203 Berlin, Germany
| | - Dirk Lassner
- Institute for Cardiac Diagnostics and Therapy (IKDT), Moltkestr. 31, D-12203 Berlin, Germany
| |
Collapse
|
40
|
Blood Biomarkers in Minor Stroke and Transient Ischemic Attack. Neurosci Bull 2016; 32:463-8. [PMID: 27250628 DOI: 10.1007/s12264-016-0038-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/05/2016] [Indexed: 01/11/2023] Open
Abstract
Minor stroke and transient ischemic attack (TIA) are common disorders with a high rate of subsequent disabling stroke, so the early recognition and management of minor stroke and TIA is of great importance. At the moment, the diagnosis of these disorders is based on neurologic deficits in a stroke-clinician's examination of the patient, supplemented by the results of acute brain imaging. However, high variability in TIA diagnosis has been reported between physicians, even trained vascular neurologists, and image-based diagnostic confirmation is not always readily available. Some patients still have ischemic events despite sustained standard secondary preventive therapy. Blood biomarkers are promising to aid in the diagnosis, risk stratification, and individual treatment of minor stroke and TIA. Some studies are being conducted in this field. This mini-review aims to highlight potential biomarkers for diagnosis and those helpful in predicting the risk of future stroke and the selection of treatment.
Collapse
|
41
|
Mitochondrial DNA mutations in neurodegeneration. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1401-11. [PMID: 26014345 DOI: 10.1016/j.bbabio.2015.05.015] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/14/2015] [Accepted: 05/17/2015] [Indexed: 12/13/2022]
Abstract
Mitochondrial dysfunction is observed in both the aging brain, and as a core feature of several neurodegenerative diseases. A central mechanism mediating this dysfunction is acquired molecular damage to mitochondrial DNA (mtDNA). In addition, inherited stable mtDNA variation (mitochondrial haplogroups), and inherited low level variants (heteroplasmy) have also been associated with the development of neurodegenerative disease and premature neural aging respectively. Herein we review the evidence for both inherited and acquired mtDNA mutations contributing to neural aging and neurodegenerative disease. This article is part of a Special Issue entitled: Mitochondrial Dysfunction in Aging.
Collapse
|
42
|
Mitochondrial DNA haplogroups and short-term neurological outcomes of ischemic stroke. Sci Rep 2015; 5:9864. [PMID: 25993529 PMCID: PMC4438613 DOI: 10.1038/srep09864] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/23/2015] [Indexed: 01/17/2023] Open
Abstract
Stroke is one of the leading causes of death and long-term disability worldwide. Mitochondrial DNA (mtDNA) is a potential contributor for the sex differences of ischemic stroke heritability. Although mtDNA haplogroups were associated with stroke onset, their impacts on stroke outcomes remain unclear. This study aimed to evaluate the impacts of mtDNA haplogroups on short-term outcomes of neurological functions in patients with ischemic stroke. A total of 303 patients were included, and their clinical data and mtDNA sequences were analyzed. Based on the changes between baseline and 14-day follow-up stroke severity, our results showed that haplogroup N9 was an independent protective factor against neurological worsening in acute ischemic stroke patients. These findings supported that mtDNA variants play a role in post-stroke neurological recovery, thus providing evidences for future pharmacological intervention in mitochondrial function.
Collapse
|
43
|
Hagen CM, Aidt FH, Havndrup O, Hedley PL, Jensen MK, Kanters JK, Pham TT, Bundgaard H, Christiansen M. Private mitochondrial DNA variants in danish patients with hypertrophic cardiomyopathy. PLoS One 2015; 10:e0124540. [PMID: 25923817 PMCID: PMC4414448 DOI: 10.1371/journal.pone.0124540] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/19/2015] [Indexed: 02/02/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic cardiac disease primarily caused by mutations in genes coding for sarcomeric proteins. A molecular-genetic etiology can be established in ~60% of cases. Evolutionarily conserved mitochondrial DNA (mtDNA) haplogroups are susceptibility factors for HCM. Several polymorphic mtDNA variants are associated with a variety of late-onset degenerative diseases and affect mitochondrial function. We examined the role of private, non-haplogroup associated, mitochondrial variants in the etiology of HCM. In 87 Danish HCM patients, full mtDNA sequencing revealed 446 variants. After elimination of 312 (69.9%) non-coding and synonymous variants, a further 109 (24.4%) with a global prevalence > 0.1%, three (0.7%) haplogroup associated and 19 (2.0%) variants with a low predicted in silico likelihood of pathogenicity, three variants: MT-TC: m.5772G>A, MT-TF: m.644A>G, and MT-CYB: m.15024G>A, p.C93Y remained. A detailed analysis of these variants indicated that none of them are likely to cause HCM. In conclusion, private mtDNA mutations are frequent, but they are rarely, if ever, associated with HCM.
Collapse
Affiliation(s)
- Christian M. Hagen
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederik H. Aidt
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Ole Havndrup
- Department of Cardiology, Roskilde Hospital, Roskilde, Denmark
| | - Paula L. Hedley
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Morten K. Jensen
- Department of Medicine B, The Heart Center, Rigshospitalet, Copenhagen, Denmark
| | - Jørgen K. Kanters
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tam T. Pham
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Medicine B, The Heart Center, Rigshospitalet, Copenhagen, Denmark
| | - Michael Christiansen
- Department of Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
44
|
Salas A, Elson JL. Mitochondrial DNA as a risk factor for false positives in case-control association studies. J Genet Genomics 2015; 42:169-72. [PMID: 25953355 DOI: 10.1016/j.jgg.2015.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/12/2015] [Accepted: 03/10/2015] [Indexed: 02/04/2023]
Affiliation(s)
- Antonio Salas
- Unidade de Xenética, Instituto de Medicina Legal, and Departamento de Anatomía Patolóxica e Ciencias Forenses, Facultad de Medicina, Universidad de Santiago de Compostela, Galicia 15782, Spain.
| | - Joanna L Elson
- Mitochondrial Research Group, Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; Centre for Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
45
|
Abstract
In saturation diving, divers stay under pressure until most of their tissues are saturated with breathing gas. Divers spend a long time in isolation exposed to increased partial pressure of oxygen, potentially toxic gases, bacteria, and bubble formation during decompression combined with shift work and long periods of relative inactivity. Hyperoxia may lead to the production of reactive oxygen species (ROS) that interact with cell structures, causing damage to proteins, lipids, and nucleic acid. Vascular gas-bubble formation and hyperoxia may lead to dysfunction of the endothelium. The antioxidant status of the diver is an important mechanism in the protection against injury and is influenced both by diet and genetic factors. The factors mentioned above may lead to production of heat shock proteins (HSP) that also may have a negative effect on endothelial function. On the other hand, there is a great deal of evidence that HSPs may also have a "conditioning" effect, thus protecting against injury. As people age, their ability to produce antioxidants decreases. We do not currently know the capacity for antioxidant defense, but it is reasonable to assume that it has a limit. Many studies have linked ROS to disease states such as cancer, insulin resistance, diabetes mellitus, cardiovascular diseases, and atherosclerosis as well as to old age. However, ROS are also involved in a number of protective mechanisms, for instance immune defense, antibacterial action, vascular tone, and signal transduction. Low-grade oxidative stress can increase antioxidant production. While under pressure, divers change depth frequently. After such changes and at the end of the dive, divers must follow procedures to decompress safely. Decompression sickness (DCS) used to be one of the major causes of injury in saturation diving. Improved decompression procedures have significantly reduced the number of reported incidents; however, data indicate considerable underreporting of injuries. Furthermore, divers who are required to return to the surface quickly are under higher risk of serious injury as no adequate decompression procedures for such situations are available. Decompression also leads to the production of endothelial microparticles that may reduce endothelial function. As good endothelial function is a documented indicator of health that can be influenced by regular exercise, regular physical exercise is recommended for saturation divers. Nowadays, saturation diving is a reasonably safe and well controlled method for working under water. Until now, no long-term impact on health due to diving has been documented. However, we still have limited knowledge about the pathophysiologic mechanisms involved. In particular we know little about the effect of long exposure to hyperoxia and microparticles on the endothelium.
Collapse
Affiliation(s)
- Alf O Brubakk
- Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | | |
Collapse
|
46
|
Fachal L, Mosquera-Miguel A, Pastor P, Ortega-Cubero S, Lorenzo E, Oterino-Durán A, Toriello M, Quintáns B, Camiña-Tato M, Sesar A, Vega A, Sobrido MJ, Salas A. No evidence of association between common European mitochondrial DNA variants in Alzheimer, Parkinson, and migraine in the Spanish population. Am J Med Genet B Neuropsychiatr Genet 2015; 168B:54-65. [PMID: 25349034 DOI: 10.1002/ajmg.b.32276] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 09/25/2014] [Indexed: 11/07/2022]
Abstract
Certain mitochondrial DNA (mtDNA) variants and haplogroups have been found to be associated with neurological disorders. Several studies have suggested that mtDNA variation could have an etiologic role in these disorders by affecting the ATP production on high-energy demanding organs, such as the brain. We have analyzed 15 mtDNA SNPs (mtSNPs) in five cohorts of cases presenting Alzheimer disease (AD), Parkinson disease (PD), and migraine, and in controls, to evaluate the role mtDNA variation in disease risk. Association tests were undertaken both for mtSNPs and mitochondrial haplogroups. No significant association was detected for any mtSNP or haplogroup in AD and PD cohorts. Two mtSNPs were associated with one migraine cohort after correcting for multiple tests, namely, T4216C and G13708A and haplogroup J (FDR q-value = 0.02; Santiago's cohort). However, this association was not confirmed in a second replication migraine series. A review of the literature reveals the existence of inconsistent findings and methodological shortcomings affecting a large proportion of mtDNA association studies on AD, PD, and migraine. A detailed inspection of the literature highlights the need for performing more rigorous methodological and statistical standards in mtDNA genetic association studies aimed to avoid false positive results of association between mtDNA variants and neurological diseases.
Collapse
Affiliation(s)
- Laura Fachal
- Fundación Pública Galega de Medicina Xenómica-SERGAS, Grupo de Medicina Xenómica, CIBERER, IDIS, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Liu Y, Zhu Q, Zhu C, Wang X, Yang J, Yin T, Gao J, Li Z, Ma Q, Guan M, Li Y, Chen Y. Systematic analysis of the clinical and biochemical characteristics of maternally inherited hypertension in Chinese Han families associated with mitochondrial. BMC Med Genomics 2014; 7:73. [PMID: 25539907 PMCID: PMC4331388 DOI: 10.1186/s12920-014-0073-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 12/16/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mitochondrial DNA mutations may be associated with cardiovascular disease, including the common cardiac vascular disease, hypertension. METHODS In this study we performed segregation analysis and systematically evaluated the entire mitochondrial genome in nine maternally inherited hypertension probands from Chinese Han families. We also performed clinical, genetic and molecular characterization of 74 maternally inherited members from these families and 216 healthy controls. RESULTS In the maternally inherited members, 12 had coronary heart disease (CHD), six had cerebrovascular disease, five had diabetes, nine had hyperlipidemia and three had renal disease. Laboratory tests showed that the sodium and potassium levels in blood of the maternally inherited members were higher than those of the control group (P < 0.01), while no differences were observed in fasting blood glucose (FBG), total cholesterol (TC), triglyceride, low density lipoprotein cholesterol (LDL-c) and creatinine levels (P > 0.05). The high density lipoprotein cholesterol (HDL-c) level of the maternally inherited members was lower than that of the control group (P = 0.04). The whole mitochondrial DNA sequence analysis revealed a total of 172 base changes, including 17 in ribosomal RNA (rRNA) genes, four in transfer RNA (tRNA) genes, and 22 amino acid substitutions. The remainder were synonymous changes or were located in non-coding regions. We identified seven amino acid changes in the nine maternally inherited hypertension families, including four mutations in ATPase6 and three in Cytb. More interestingly, tRNA(Ser(UCN)) 7492 T > C was absent in controls and was present in <1% of 2704 mtDNAs, indicating potential functional significance. CONCLUSIONS This study showed that mutations in mtDNA may contribute to the pathogenesis of hypertension in these Chinese Han families. In the near future, identification of additional mtDNA mutations may indicate further candidate genes for hypertension.
Collapse
Affiliation(s)
- Yuqi Liu
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Qinglei Zhu
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Chao Zhu
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Xueping Wang
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Jie Yang
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Tong Yin
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Jinliao Gao
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Zongbin Li
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
| | - Qinghua Ma
- Department of Cardiology, Yishui Center Hospital of Shandong Province, Yishui, Shandong, China.
| | - Minxin Guan
- Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical College, Wenzhou, Zhejiang, China.
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yang Li
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
- Cardiology Department of General Hospital of People's Liberation Army, Institute of Geriatric Cardiology, No. 28 Fuxing Road, Hai dian District, Beijing, 100853, PR of China.
| | - Yundai Chen
- Cardiology Department, Chinese PLA General Hospital, Beijing, China.
- Cardiology Department of General Hospital of People's Liberation Army, Institute of Geriatric Cardiology, No. 28 Fuxing Road, Hai dian District, Beijing, 100853, PR of China.
| |
Collapse
|
48
|
Yang D, Wang Q, Shi Y, Fan Y, Zheng HX, Song G, Feng Q, Zheng H, He Y. Mitochondrial DNA haplogroup D4b is a protective factor for ischemic stroke in Chinese Han population. Mol Genet Genomics 2014; 289:1241-6. [PMID: 25092472 DOI: 10.1007/s00438-014-0884-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 06/28/2014] [Indexed: 01/01/2023]
Abstract
Mitochondrial DNA (mtDNA) haplogroups affect the assembly and stability of the mitochondrial respiratory chain, which is potentially related to susceptibility to ischemic stroke (IS). However, the role of mtDNA in IS has not been comprehensively studied. The purpose of this study was to explore whether mtDNA polymorphisms and haplogroups are involved in the etiology of IS in the Chinese Han population. We recruited 200 patients with IS and 200 matched controls and genotyped them for 18 mtDNA single nucleotide polymorphisms defining the major Eastern Asian haplogroups by SNaPshot minisequencing. We also sequenced the hypervariable segment I (HVS-I), position 16051-16400. The prevalence of haplogroup D4b was significantly lower in IS patients than in healthy controls (0 and 8 %, respectively, corrected P = 2 × 10(-5), odds ratio = 0.028, 95 % confidence interval = 0.002-0.468).The positive association between haplogroup D4b and IS may be related to the protective effect of haplogroup D4b against oxidative damage, which decreases the risk of IS. Our study provides the first evidence that haplogroup D4b is a potential genetic protective factor for IS in the Chinese Han population.
Collapse
Affiliation(s)
- Dongzhi Yang
- School of life sciences of Zhengzhou University, Zhengzhou, 450052, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhang W, Tang J, Zhang AM, Peng MS, Xie HB, Tan L, Xu L, Zhang YP, Chen X, Yao YG. A Matrilineal Genetic Legacy from the Last Glacial Maximum Confers Susceptibility to Schizophrenia in Han Chinese. J Genet Genomics 2014; 41:397-407. [DOI: 10.1016/j.jgg.2014.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/16/2014] [Accepted: 05/21/2014] [Indexed: 10/25/2022]
|
50
|
Recent mitochondrial DNA mutations increase the risk of developing common late-onset human diseases. PLoS Genet 2014; 10:e1004369. [PMID: 24852434 PMCID: PMC4031051 DOI: 10.1371/journal.pgen.1004369] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/24/2014] [Indexed: 02/01/2023] Open
Abstract
Mitochondrial DNA (mtDNA) is highly polymorphic at the population level, and specific mtDNA variants affect mitochondrial function. With emerging evidence that mitochondrial mechanisms are central to common human diseases, it is plausible that mtDNA variants contribute to the “missing heritability” of several complex traits. Given the central role of mtDNA genes in oxidative phosphorylation, the same genetic variants would be expected to alter the risk of developing several different disorders, but this has not been shown to date. Here we studied 38,638 individuals with 11 major diseases, and 17,483 healthy controls. Imputing missing variants from 7,729 complete mitochondrial genomes, we captured 40.41% of European mtDNA variation. We show that mtDNA variants modifying the risk of developing one disease also modify the risk of developing other diseases, thus providing independent replication of a disease association in different case and control cohorts. High-risk alleles were more common than protective alleles, indicating that mtDNA is not at equilibrium in the human population, and that recent mutations interact with nuclear loci to modify the risk of developing multiple common diseases. There is a growing body of evidence indicating that mitochondrial dysfunction, a result of genetic variation in the mitochondrial genome, is a critical component in the aetiology of a number of complex traits. Here, we take advantage of recent technical and methodological advances to examine the role of common mitochondrial DNA variants in several complex diseases. By examining over 50,000 individuals, from 11 different diseases we show that mitochondrial DNA variants can both increase or decrease an individual's risk of disease, replicating and expanding upon several previously reported studies. Moreover, by analysing several large disease groups in tandem, we are able to show a commonality of association, with the same mitochondrial DNA variants associated with several distinct disease phenotypes. These shared genetic associations implicate a shared underlying functional effect, likely changing cellular energy, which manifests as distinct phenotypes. Our study confirms the important role that mitochondrial DNA variation plays on complex traits and additionally supports the utility of a GWAS-based approach for analysing mitochondrial genetics.
Collapse
|