1
|
Fernandez-Gonzalez SM, Sucasas-Alonso A, Balboa-Barreiro V, Rego-Perez I, Avila-Alvarez A. Mitochondrial DNA haplogroups and circulating cell-free mitochondrial DNA as biomarkers of bronchopulmonary dysplasia. Pediatr Res 2025:10.1038/s41390-025-04052-7. [PMID: 40247115 DOI: 10.1038/s41390-025-04052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/06/2025] [Accepted: 03/02/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Recognizing which premature infants are at higher risk of developing BPD/death is a challenge in neonatology. The aims of our study are to identify mitochondrial haplogroups and quantify circulating cell-free mitochondrial DNA (ccf-mtDNA) levels in very preterm infants at risk of bronchopulmonary dysplasia (BPD) or death and explore the relationship between these variables and the development of BPD/death. METHODS Single-center prospective cohort study including preterm infants of ≤32 weeks gestational age (GA) and birth weight ≤1500 g. Clinical variables, mitochondrial haplogroups, and ccf-mtDNA levels were determined. Subsequently, diagnosis and staging of BPD/death were performed, and groups were compared. RESULTS The population consisted of 107 newborns (mean GA 28.73 ± 2 weeks; mean birth weight 1,121 ± 332 g). A total of 44 patients (41.1%) presented the outcome of BPD/death without differences in haplogroup distribution and ccf-mtDNA levels between those who survived without BPD (controls). Variables independently associated with BPD/death included GA (p < 0.001; OR = 0.36 [95%CI 0.23-0.5]), birth weight (p < 0.001; OR = 0.99 [95%CI 0.99-0.99]), maximum FiO2 in the delivery room (p = 0.001; OR = 1.07 [95%CI 1.03-1.12]), hours on mechanical ventilation (p = 0.02; OR 1.02 [95%CI 1.00-1.02]), and postnatal corticosteroids (p < 0.001; OR = 47.12 [95%CI = 5.98-371.1]). CONCLUSION This is the first study to characterize mtDNA haplogroups and ccf-mtDNA in very preterm infants at risk of BPD/death. None of the mitochondrial variables studied were associated with BPD/death. Further research is needed to elucidate the role of mtDNA in BPD. IMPACT STATEMENT Despite advances in perinatal care, bronchopulmonary dysplasia continues to be the most common chronic pulmonary morbidity associated with prematurity. Prediction of BPD in early stages is crucial to improve BPD rates, but this remains a major challenge in neonatal units. Given that mitochondria play an important role in the inflammatory and oxidative stress responses, we aimed to explore the relationship between mitochondrial haplogroups, circulating cell-free mitochondrial DNA levels, and BPD. This is the first work carried out in very preterm infants where mitochondrial haplogroups and the levels ccf-mtDNA are investigated with the intention of discovering a new biomarker for BPD.
Collapse
Affiliation(s)
| | - Andrea Sucasas-Alonso
- Neonatology Department, Complexo Hospitalario Universitario A Coruña (CHUAC), A Coruña, Spain
| | - Vanesa Balboa-Barreiro
- Biostatistics and Epidemiology Unit, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Ignacio Rego-Perez
- Grupo de Investigación en Reumatología, Unidad de Genómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain
| | - Alejandro Avila-Alvarez
- Neonatology Department, Complexo Hospitalario Universitario A Coruña (CHUAC), A Coruña, Spain.
- Grupo de Investigación en Pediatría. Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario, Universitario A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), A Coruña, Spain.
| |
Collapse
|
2
|
Atabekov T, Korepanov V, Krivolapov S, Khlynin M, Afanasiev S, Golubenko M, Batalov R, Popov S. Mitochondrial DNA Polymorphisms of Peripheral Blood Mononuclear Cells Associated with Sustained Ventricular Tachycardia in Patients with Cardioverter-Defibrillator Implantation Indications. Rev Cardiovasc Med 2025; 26:26744. [PMID: 40160580 PMCID: PMC11951477 DOI: 10.31083/rcm26744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 04/02/2025] Open
Abstract
Background Mitochondrial dysfunction in myocardium cells has been implicated in arrhythmogenesis, including ventricular tachycardia (VT). A carriage of point mitochondrial DNA (mtDNA) polymorphisms may contribute to the risk of certain arrhythmias. Therefore, it is hypothesized that mtDNA genotype could predict the risk of sustained VT (SVT). We aimed to explore whether specific mtDNA polymorphisms of peripheral blood mononuclear cells (PBMC) can serve as biomarkers for predicting the risk of SVT in patients with indications for an implantable cardioverter-defibrillator (ICD). Methods A total of 122 patients with ICD implantation indications who underwent transthoracic echocardiography (TTE) were enrolled in the study. Total DNA from PBMC was isolated using the phenol-chloroform extraction method. Genotyping of mtDNA polymorphisms A2706G, G3010A and G9055A was performed using restriction fragment length polymorphism analysis. Correlations between clinical parameters and mtDNA polymorphisms with SVT registered prior to ICD implantation were evaluated. Based on our data, we developed a risk model for SVT. Results Prior to ICD implantation, 70 (56.6%) patients had SVT (1st group) and 52 (43.4%) patients did not have SVT (2nd group). Patients with SVT were significantly older than patients without SVT (66.9 ± 9.9 year vs. 59.5 ± 10.6 year, p < 0.001), had a lower value estimated glomerular filtration rate (eGFR) (65.7 ± 19.7 mL/min/1.73 m2 vs. 77.9 ± 16.1 mL/min/1.73 m2, p < 0.001) and less frequently had A2706G mtDNA polymorphism (55.7% vs. 76.9%, p = 0.015). According to the multivariable logistic regression, age (odds ratio (OR) = 1.055, 95% confidence interval (CI) 1.009-1.103, p = 0.017), eGFR (OR = 0.974, 95% CI 0.949-0.999, p = 0.041) and absence of A2706G mtDNA polymorphism (OR = 0.335, 95% CI 0.141-0.797, p = 0.013) were independently associated with the SVT. We constructed a logistic equation with calculation of the cut-off value. The discriminative ability of the receiver operating characteristic curve (area under the curve) was 0.761 (95% confidence interval 0.675-0.833; sensitivity 65.71%; specificity 76.92%). Conclusions In patients with ICD implantation indications, a carriage of mtDNA polymorphism A2706G is associated with SVT. Our risk model including age, eGFR and absence of A2706G mtDNA substitution was able to distinguish patients with SVT. Further investigations of their predictive significance are warranted. Clinical Trial Registration NCT03667989 (https://clinicaltrials.gov/study/NCT03667989).
Collapse
Affiliation(s)
- Tariel Atabekov
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Viacheslav Korepanov
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Sergey Krivolapov
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Mikhail Khlynin
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Sergey Afanasiev
- Laboratory of Molecular and Cellular Pathology and Gene Diagnostics, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Maria Golubenko
- Laboratory for Population Genetics, Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 634050 Tomsk, Russia
| | - Roman Batalov
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Sergey Popov
- Department of Surgical Arrhythmology and Cardiac Pacing, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| |
Collapse
|
3
|
Kuan SW, Chua KH, Tan EW, Tan LK, Loch A, Kee BP. Whole mitochondrial genome sequencing of Malaysian patients with cardiomyopathy. PeerJ 2022; 10:e13265. [PMID: 35441061 PMCID: PMC9013480 DOI: 10.7717/peerj.13265] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/23/2022] [Indexed: 01/13/2023] Open
Abstract
Cardiomyopathy (CMP) constitutes a diverse group of myocardium diseases affecting the pumping ability of the heart. Genetic predisposition is among the major factors affecting the development of CMP. Globally, there are over 100 genes in autosomal and mitochondrial DNA (mtDNA) that have been reported to be associated with the pathogenesis of CMP. However, most of the genetic studies have been conducted in Western countries, with limited data being available for the Asian population. Therefore, this study aims to investigate the mutation spectrum in the mitochondrial genome of 145 CMP patients in Malaysia. Long-range PCR was employed to amplify the entire mtDNA, and whole mitochondrial genome sequencing was conducted on the MiSeq platform. Raw data was quality checked, mapped, and aligned to the revised Cambridge Reference Sequence (rCRS). Variants were named, annotated, and filtered. The sequencing revealed 1,077 variants, including 18 novel and 17 CMP and/or mitochondrial disease-associated variants after filtering. In-silico predictions suggested that three of the novel variants (m.8573G>C, m.11916T>A and m.11918T>G) in this study are potentially pathogenic. Two confirmed pathogenic variants (m.1555A>G and m.11778G>A) were also found in the CMP patients. The findings of this study shed light on the distribution of mitochondrial mutations in Malaysian CMP patients. Further functional studies are required to elucidate the role of these variants in the development of CMP.
Collapse
Affiliation(s)
- Sheh Wen Kuan
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - E-Wei Tan
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Lay Koon Tan
- National Heart Institute, Kuala Lumpur, Malaysia
| | - Alexander Loch
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Boon Pin Kee
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Abstract
PROPOSE OF REVIEW To summarize the evidence that suggests that osteoarthritis (OA) is a mitochondrial disease. RECENT FINDINGS Mitochondrial dysfunction together with mtDNA damage could contribute to cartilage degradation via several processes such as: (1) increased apoptosis; (2) decreased autophagy; (3) enhanced inflammatory response; (4) telomere shortening and increased senescence chondrocytes; (5) decreased mitochondrial biogenesis and mitophagy; (6) increased cartilage catabolism; (7) increased mitochondrial fusion leading to further reactive oxygen species production; and (8) impaired metabolic flexibility. SUMMARY Mitochondria play an important role in some events involved in the pathogenesis of OA, such as energy production, the generation of reactive oxygen and nitrogen species, apoptosis, authophagy, senescence and inflammation. The regulation of these processes in the cartilage is at least partially controlled by retrograde regulation from mitochondria and mitochondrial genetic variation. Retrograde regulation through mitochondrial haplogroups exerts a signaling control over the nuclear epigenome, which leads to the modulation of nuclear genes, cellular functions and development of OA. All these data suggest that OA could be considered a mitochondrial disease as well as other complex chronic disease as cancer, cardiovascular and neurologic diseases.
Collapse
|
5
|
Mitophagy: At the heart of mitochondrial quality control in cardiac aging and frailty. Exp Gerontol 2021; 153:111508. [PMID: 34358665 DOI: 10.1016/j.exger.2021.111508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular disease is highly prevalent among older adults and poses a huge burden on morbidity, disability, and mortality. The age-related increased vulnerability of the cardiovascular system towards stressors is a pathophysiological trait of cardiovascular disease. This has been associated with a progressive deterioration of blood vessels and decline in heart function during aging. Cardiomyocytes rely mostly on oxidative metabolism for deploying their activities and mitochondrial metabolism is crucial to this purpose. Dysmorphic, inefficient, and oxidant-producing mitochondria have been identified in aged cardiomyocytes in association with cardiac structural and functional alterations. These aberrant organelles are thought to arise from inefficient mitochondrial quality control, which has therefore been place in the spotlight as a relevant mechanism of cardiac aging. As a result of alterations in mitochondrial quality control and redox dyshomeostasis, mitochondrial damage accumulates and contributes to cardiac frailty. Herein, we discuss the contribution of defective mitochondrial quality control pathways to cardiac frailty. Emerging findings pointing towards the exploitation of these pathways as therapeutic targets against cardiac aging and cardiovascular disease will also be illustrated.
Collapse
|
6
|
Phadwal K, Vrahnas C, Ganley IG, MacRae VE. Mitochondrial Dysfunction: Cause or Consequence of Vascular Calcification? Front Cell Dev Biol 2021; 9:611922. [PMID: 33816463 PMCID: PMC8010668 DOI: 10.3389/fcell.2021.611922] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are crucial bioenergetics powerhouses and biosynthetic hubs within cells, which can generate and sequester toxic reactive oxygen species (ROS) in response to oxidative stress. Oxidative stress-stimulated ROS production results in ATP depletion and the opening of mitochondrial permeability transition pores, leading to mitochondria dysfunction and cellular apoptosis. Mitochondrial loss of function is also a key driver in the acquisition of a senescence-associated secretory phenotype that drives senescent cells into a pro-inflammatory state. Maintaining mitochondrial homeostasis is crucial for retaining the contractile phenotype of the vascular smooth muscle cells (VSMCs), the most prominent cells of the vasculature. Loss of this contractile phenotype is associated with the loss of mitochondrial function and a metabolic shift to glycolysis. Emerging evidence suggests that mitochondrial dysfunction may play a direct role in vascular calcification and the underlying pathologies including (1) impairment of mitochondrial function by mineral dysregulation i.e., calcium and phosphate overload in patients with end-stage renal disease and (2) presence of increased ROS in patients with calcific aortic valve disease, atherosclerosis, type-II diabetes and chronic kidney disease. In this review, we discuss the cause and consequence of mitochondrial dysfunction in vascular calcification and underlying pathologies; the role of autophagy and mitophagy pathways in preventing mitochondrial dysfunction during vascular calcification and finally we discuss mitochondrial ROS, DRP1, and HIF-1 as potential novel markers and therapeutic targets for maintaining mitochondrial homeostasis in vascular calcification.
Collapse
Affiliation(s)
- Kanchan Phadwal
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| | - Christina Vrahnas
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Ian G. Ganley
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Vicky E. MacRae
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
7
|
Roselló-Díez E, Hove-Madsen L, Pérez-Grijalba V, Muñoz-Guijosa C, Artigas V, Maria Padró J, Domínguez-Garrido E. Mitochondrial genetic effect on atrial fibrillation: A case-control study. Mitochondrion 2020; 56:15-24. [PMID: 33171269 DOI: 10.1016/j.mito.2020.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/25/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022]
Abstract
Atrial fibrillation (AF) is a common arrhythmia in the general population and following cardiac surgery. The influence of mitochondrial genomics on AF pathogenesis is not fully understood. We analyzed mitochondrial variables from 78 human atrial samples collected from cardiac surgeries in the following groups: 1) permanent preoperative AF; 2) preoperative sinus rhythm (SR) with postoperative AF; and 3) pre-/postoperative SR. Haplogroup H appeared offer protection against, and haplogroup U predispose to permanent AF. mtDNA content was higher in group 2 than in 3. These findings contribute to a better understanding of the influence of mitochondria on AF pathogenesis.
Collapse
Affiliation(s)
- Elena Roselló-Díez
- Department of Cardiac Surgery, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, 167 Sant Antoni Maria Claret Street, 08025 Barcelona, Spain.
| | - Leif Hove-Madsen
- Cardiovascular Research Centre (CSIC) and CIBERCV, 167 Sant Antoni Maria Claret Street, 08025 Barcelona, Spain
| | - Virginia Pérez-Grijalba
- Molecular Diagnostic Unit, Fundación Rioja Salud (FRS), 98 Piqueras Street, 26006 Logroño, Spain
| | - Christian Muñoz-Guijosa
- Department of Cardiac Surgery, Universitat Autònoma de Barcelona, Hospital Universitario Germans Trias i Pujol, Canyet Road, 08916 Badalona, Spain
| | - Vicenç Artigas
- Department of General and Digestive Surgery, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, 167 Sant Antoni Maria Claret Street, 08025 Barcelona, Spain
| | - Josep Maria Padró
- Department of Cardiac Surgery, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, 167 Sant Antoni Maria Claret Street, 08025 Barcelona, Spain
| | - Elena Domínguez-Garrido
- Molecular Diagnostic Unit, Fundación Rioja Salud (FRS), 98 Piqueras Street, 26006 Logroño, Spain
| |
Collapse
|
8
|
Bayona-Bafaluy MP, Iglesias E, López-Gallardo E, Emperador S, Pacheu-Grau D, Labarta L, Montoya J, Ruiz-Pesini E. Genetic aspects of the oxidative phosphorylation dysfunction in dilated cardiomyopathy. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 786:108334. [PMID: 33339579 DOI: 10.1016/j.mrrev.2020.108334] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/27/2022]
Abstract
Dilated cardiomyopathy is a frequent and extremely heterogeneous medical condition. Deficits in the oxidative phosphorylation system have been described in patients suffering from dilated cardiomyopathy. Hence, mutations in proteins related to this biochemical pathway could be etiological factors for some of these patients. Here, we review the clinical phenotypes of patients harboring pathological mutations in genes related to the oxidative phosphorylation system, either encoded in the mitochondrial or in the nuclear genome, presenting with dilated cardiomyopathy. In addition to the clinical heterogeneity of these patients, the large genetic heterogeneity has contributed to an improper allocation of pathogenicity for many candidate mutations. We suggest criteria to avoid incorrect assignment of pathogenicity to newly found mutations and discuss possible therapies targeting the oxidative phosphorylation function.
Collapse
Affiliation(s)
- M Pilar Bayona-Bafaluy
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza. C/ Miguel Servet, 177. 50013, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13., 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Eldris Iglesias
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza. C/ Miguel Servet, 177. 50013, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13., 50009, Zaragoza, Spain.
| | - Ester López-Gallardo
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza. C/ Miguel Servet, 177. 50013, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13., 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Sonia Emperador
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza. C/ Miguel Servet, 177. 50013, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13., 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - David Pacheu-Grau
- Department of Cellular Biochemistry, University Medical Center, Georg-August University,Humboldtalle, 23., 37073, Göttingen, Germany.
| | - Lorenzo Labarta
- Unidad de Cuidados Intensivos, Hospital San Jorge, Av. Martínez de Velasco, 36., 22004, Huesca, Spain.
| | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza. C/ Miguel Servet, 177. 50013, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13., 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza. C/ Miguel Servet, 177. 50013, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13., 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Fundación ARAID, Av. de Ranillas, 1-D., 50018, Zaragoza, Spain.
| |
Collapse
|
9
|
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: 0.2] [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
|
10
|
Kytövuori L, Junttila J, Huikuri H, Keinänen-Kiukaanniemi S, Majamaa K, Martikainen MH. Mitochondrial DNA variation in sudden cardiac death: a population-based study. Int J Legal Med 2019; 134:39-44. [PMID: 31152278 PMCID: PMC6949201 DOI: 10.1007/s00414-019-02091-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/22/2019] [Indexed: 11/26/2022]
Abstract
Cardiomyopathy and cardiac conduction defects are common manifestations of mitochondrial disease. Previous studies suggest that clinically asymptomatic individuals harbouring pathogenic mitochondrial DNA (mtDNA) mutations in the cardiac muscle may have sudden cardiac death (SCD) as the first manifestation of mitochondrial disease. We investigated the contribution of pathogenic mtDNA point mutations and mtDNA haplogroups in cardiac muscle in a cohort of 280 Finnish subjects that had died from non-ischaemic SCD with the median age of death at 59 years and in 537 population controls. We did not find any common or novel pathogenic mutations, but the frequency of haplogroup H1 was higher in the SCD subjects than that in 537 population controls (odds ratio: 1.76, confidence interval 95%: 1.02-3.04). We conclude that, at the population level, pathogenic point mutations in mtDNA do not contribute to non-ischaemic SCD, but natural variation may modify the risk.
Collapse
Affiliation(s)
- Laura Kytövuori
- Research Unit of Clinical Neuroscience, University of Oulu, PO Box 5000, 90014 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Neurology, Oulu University Hospital, PO Box 20, 90029 Oulu, Finland
| | - Juhani Junttila
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Research Unit of Internal Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland
| | - Heikki Huikuri
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Research Unit of Internal Medicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland
| | - Sirkka Keinänen-Kiukaanniemi
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Center for Life Course Health Research, University of Oulu, PO Box 5000, 90014 Oulu, Finland
- Healthcare and Social Services of Selänne, Pyhäjärvi, Finland
| | - Kari Majamaa
- Research Unit of Clinical Neuroscience, University of Oulu, PO Box 5000, 90014 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Neurology, Oulu University Hospital, PO Box 20, 90029 Oulu, Finland
| | - Mika H. Martikainen
- Division of Clinical Neurosciences, University of Turku and Turku University Hospital, Kiinamyllynkatu 4-8, 20520 Turku, Finland
| |
Collapse
|
11
|
Govindaraj P, Rani B, Sundaravadivel P, Vanniarajan A, Indumathi KP, Khan NA, Dhandapany PS, Rani DS, Tamang R, Bahl A, Narasimhan C, Rakshak D, Rathinavel A, Premkumar K, Khullar M, Thangaraj K. Mitochondrial genome variations in idiopathic dilated cardiomyopathy. Mitochondrion 2019; 48:51-59. [PMID: 30910572 DOI: 10.1016/j.mito.2019.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 03/19/2019] [Indexed: 12/22/2022]
Abstract
Idiopathic dilated cardiomyopathy (DCM) is a structural heart disease with strong genetic background. The aim of this study was to assess the role of mitochondrial DNA (mtDNA) variations and haplogroups in Indian DCM patients. Whole mtDNA analysis of 221 DCM patients revealed 48 novel, 42 disease-associated and 97 private variations. The frequency of reported variations associated with hearing impairment, DEAF, SNHL and LHON are significantly high in DCM patients than controls. Haplogroups H and HV were over represented in DCM than controls. Functional analysis of two private variations (m.8812A>G & m.10320G>A) showed decrease in mitochondrial functions, suggesting the role of mtDNA variations in DCM.
Collapse
Affiliation(s)
- Periyasamy Govindaraj
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India; Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Bindu Rani
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | | | - K P Indumathi
- Department of Biochemistry, Kongu Arts and Science College, Erode, India
| | | | - Perundurai S Dhandapany
- Centre for Cardiovascular Biology and disease, Institute of Stem Cell Biology and Regenerative Medicine(inStem), Bengaluru, India; The Knight Cardiovascular Institute, Departments of Medicine, Molecular and Medical Genetics, Oregon Health and Science University, Portland, USA
| | - Deepa Selvi Rani
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Rakesh Tamang
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Ajay Bahl
- Department of Cardiology, PGIMER, Chandigarh, India
| | | | | | - Andiappan Rathinavel
- Department of Cardio-Thoracic Surgery, Madurai Medical College & Government Rajaji hospital, Madurai, India
| | - Kumpati Premkumar
- Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | |
Collapse
|
12
|
Serrano-Teruel ME, Garcia-Vieites M, Rego-Perez I, Domenech-Garcia N, Blanco-Garcia F, Cuenca-Castillo JJ, Bautista-Hernandez V. Mitochondrial DNA haplogroups influence the risk of aortic stenosis. Asian Cardiovasc Thorac Ann 2018; 27:5-10. [PMID: 30409026 DOI: 10.1177/0218492318813220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIM The underlying pathophysiologic mechanisms of aortic stenosis are not clear. Mitochondrial dysfunction plays a role in many pathological conditions including cardiac diseases. We aimed to analyze the mitochondrial DNA haplogroups in a group of patients undergoing valve replacement surgery due to severe aortic stenosis. METHODS Mitochondrial DNA haplogroups were assessed in 176 patients with severe aortic stenosis and 308 control subjects. Cardiovascular risk factors and demographics were similar in both groups. RESULTS Patients carrying haplogroup Uk had a lower risk of developing aortic stenosis, especially compared to patients carrying haplogroup H (odds ratio = 0.507; 95% confidence interval: 0.270-0.952, p = 0.035). CONCLUSIONS Mitochondrial DNA haplogroups could be involved in the development of severe aortic stenosis. Specifically, haplogroup H could be a risk factor and Uk a protective factor for severe aortic stenosis in a population from Spain.
Collapse
Affiliation(s)
- Maria E Serrano-Teruel
- 1 Congenital & Structural Heart Disease, A Coruña Biomedical Research Institute (INIBIC) A Coruña, Spain
| | - Maria Garcia-Vieites
- 1 Congenital & Structural Heart Disease, A Coruña Biomedical Research Institute (INIBIC) A Coruña, Spain.,2 Department of Cardiovascular Surgery. University Hospital Complex A Coruña (CHUAC), A Coruña, Spain
| | - Ignacio Rego-Perez
- 3 Genomic Group. A Coruña Biomedical Research Institute (INIBIC), A Coruña, Spain
| | | | | | - Jose J Cuenca-Castillo
- 2 Department of Cardiovascular Surgery. University Hospital Complex A Coruña (CHUAC), A Coruña, Spain
| | - Victor Bautista-Hernandez
- 1 Congenital & Structural Heart Disease, A Coruña Biomedical Research Institute (INIBIC) A Coruña, Spain.,2 Department of Cardiovascular Surgery. University Hospital Complex A Coruña (CHUAC), A Coruña, Spain
| |
Collapse
|
13
|
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
|
14
|
Abstract
PURPOSE OF REVIEW Cardiovascular disease (CVD) is responsible for more morbidity and mortality worldwide than any other ailment. Strategies for reducing CVD prevalence must involve identification of individuals at high risk for these diseases, and the prevention of its initial development. Such preventive efforts are currently limited by an incomplete understanding of the genetic determinants of CVD risk. In this review, evidence for the involvement of inherited mitochondrial mutations in development of CVD is examined. RECENT FINDINGS Several forms of CVD have been documented in the presence of pathogenic mitochondrial DNA (mtDNA) mutations, both in isolation and as part of larger syndromes. Other 'natural' mtDNA polymorphisms not overtly tied to any pathology have also been associated with alterations in mitochondrial function and individual risk for CVD, but until very recently these studies have been merely correlative. Fortunately, novel animal models are now allowing investigators to define a causal relationship between inherited 'natural' mtDNA polymorphisms, and cardiovascular function and pathology. SUMMARY Cardiovascular involvement is highly prevalent among patients with pathogenic mtDNA mutations. The relationship between CVD susceptibility and 'natural' mtDNA polymorphisms requires further investigation, but will be aided in the near future by several novel experimental models.
Collapse
Affiliation(s)
- Alexander W. Bray
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham
| | - Scott W. Ballinger
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham
| |
Collapse
|
15
|
Generating Rho-0 Cells Using Mesenchymal Stem Cell Lines. PLoS One 2016; 11:e0164199. [PMID: 27764131 PMCID: PMC5072612 DOI: 10.1371/journal.pone.0164199] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/21/2016] [Indexed: 12/28/2022] Open
Abstract
Introduction The generation of Rho-0 cells requires the use of an immortalization process, or tumor cell selection, followed by culture in the presence of ethidium bromide (EtBr), incurring the drawbacks its use entails. The purpose of this work was to generate Rho-0 cells using human mesenchymal stem cells (hMSCs) with reagents having the ability to remove mitochondrial DNA (mtDNA) more safely than by using EtBr. Methodology Two immortalized hMSC lines (3a6 and KP) were used; 143B.TK-Rho-0 cells were used as reference control. For generation of Rho-0 hMSCs, cells were cultured in medium supplemented with each tested reagent. Total DNA was isolated and mtDNA content was measured by real-time polymerase chain reaction (PCR). Phenotypic characterization and gene expression assays were performed to determine whether 3a6 Rho-0 hMSCs maintain the same stem properties as untreated 3a6 hMSCs. To evaluate whether 3a6 Rho-0 hMSCs had a phenotype similar to that of 143B.TK-Rho-0 cells, in terms of reactive oxygen species (ROS) production, apoptotic levels and mitochondrial membrane potential (Δψm) were measured by flow cytometry and mitochondrial respiration was evaluated using a SeaHorse XFp Extracellular Flux Analyzer. The differentiation capacity of 3a6 and 3a6 Rho-0 hMSCs was evaluated using real-time PCR, comparing the relative expression of genes involved in osteogenesis, adipogenesis and chondrogenesis. Results The results showed the capacity of the 3a6 cell line to deplete its mtDNA and to survive in culture with uridine. Of all tested drugs, Stavudine (dt4) was the most effective in producing 3a6-Rho cells. The data indicate that hMSC Rho-0 cells continue to express the characteristic MSC cell surface receptor pattern. Phenotypic characterization showed that 3a6 Rho-0 cells resembled 143B.TK-Rho-0 cells, indicating that hMSC Rho-0 cells are Rho-0 cells. While the adipogenic capability was higher in 3a6 Rho-0 cells than in 3a6 cells, the osteogenic and chondrogenic capacities were lower. Conclusion Among the drugs and conditions tested, the use of d4t was the best option for producing Rho-0 cells from hMSCs. Rho-0 cells are useful for studying the role of mitochondria in hMSC differentiation.
Collapse
|
16
|
Thaker K, Chwa M, Atilano SR, Coskun P, Cáceres-Del-Carpio J, Udar N, Boyer DS, Jazwinski SM, Miceli MV, Nesburn AB, Kuppermann BD, Kenney MC. Increased expression of ApoE and protection from amyloid-beta toxicity in transmitochondrial cybrids with haplogroup K mtDNA. Neurobiol Dis 2016; 93:64-77. [PMID: 27109188 DOI: 10.1016/j.nbd.2016.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/13/2016] [Accepted: 04/20/2016] [Indexed: 01/09/2023] Open
Abstract
Mitochondrial (mt) DNA haplogroups, defined by specific single nucleotide polymorphism (SNP) patterns, represent populations of diverse geographic origins and have been associated with increased risk or protection of many diseases. The H haplogroup is the most common European haplogroup while the K haplogroup is highly associated with the Ashkenazi Jewish population. Transmitochondrial cybrids (cell lines with identical nuclei, but mtDNA from either H (n=8) or K (n=8) subjects) were analyzed by the Seahorse flux analyzer, quantitative polymerase chain reaction (Q-PCR) and immunohistochemistry (IHC). Cybrids were treated with amyloid-β peptides and cell viabilities were measured. Other cybrids were demethylated with 5-aza-2'-deoxycytidine (5-aza-dC) and expression levels for APOE and NFkB2 were measured. Results show K cybrids have (a) significantly lower mtDNA copy numbers, (b) higher expression levels for MT-DNA encoded genes critical for oxidative phosphorylation, (c) lower Spare Respiratory Capacity, (d) increased expression of inhibitors of the complement pathway and important inflammasome-related genes; and (e) significantly higher levels of APOE transcription that were independent of methylation status. After exposure to amyloid-β1-42 peptides (active form), H haplogroup cybrids demonstrated decreased cell viability compared to those treated with amyloid-β42-1 (inactive form) (p<0.0001), while this was not observed in the K cybrids (p=0.2). K cybrids had significantly higher total global methylation levels and differences in expression levels for two acetylation genes and four methylation genes. Demethylation with 5-aza-dC altered expression levels for NFkB2, while APOE transcription patterns were unchanged. Our findings support the hypothesis that mtDNA-nuclear retrograde signaling may mediate expression levels of APOE, a key factor in many age-related diseases. Future studies will focus on identification of the mitochondrial-nuclear retrograde signaling mechanism(s) contributing to these mtDNA-mediated differences.
Collapse
Affiliation(s)
- Kunal Thaker
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, United States
| | - Marilyn Chwa
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, United States
| | - Shari R Atilano
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, United States
| | - Pinar Coskun
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, United States
| | | | - Nitin Udar
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, United States
| | - David S Boyer
- Retina-Vitreous Associates Medical Group, Beverly Hills, CA, United States
| | - S Michal Jazwinski
- Tulane Center for Aging and Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Michael V Miceli
- Tulane Center for Aging and Department of Medicine, Tulane University, New Orleans, LA, United States
| | - Anthony B Nesburn
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, United States; Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Baruch D Kuppermann
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, United States
| | - M Cristina Kenney
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, United States; Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, United States.
| |
Collapse
|
17
|
St John JC. Mitochondrial DNA copy number and replication in reprogramming and differentiation. Semin Cell Dev Biol 2016; 52:93-101. [PMID: 26827792 DOI: 10.1016/j.semcdb.2016.01.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 01/11/2016] [Accepted: 01/21/2016] [Indexed: 11/26/2022]
Abstract
Until recently, it was thought that the role of the mitochondrial genome was confined to encoding key proteins that generate ATP through the process of oxidative phosphorylation in the electron transfer chain. However, with increasing new evidence, it is apparent that the mitochondrial genome has a major role to play in a number of diseases and phenotypes. For example, mitochondrial variants and copy number have been implicated in the processes of fertilisation outcome and development and the onset of tumorigenesis. On the other hand, mitochondrial DNA (mtDNA) haplotypes have been implicated in a variety of diseases and most likely account for the adaptation that our ancestors achieved in order that they were fit for their environments. The mechanisms, which enable the mitochondrial genome to either protect or promote the disease phenotype, require further elucidation. However, there appears to be significant 'crosstalk' between the chromosomal and mitochondrial genomes that enable this to take place. One such mechanism is the regulation of DNA methylation by mitochondrial DNA, which is often perturbed in reprogrammed cells that have undergone dedifferentiation and affects mitochondrial DNA copy number. Furthermore, it appears that the mitochondrial genome interacts with the chromosomal genome to regulate the transcription of key genes at certain stages during development. Additionally, the mitochondrial genome can accumulate a series of mtDNA variants, which can lead to diseases such as cancer. It is likely that a combination of certain mitochondrial variants and aberrant patterns of mtDNA copy number could indeed account for many diseases that have previously been unaccounted for. This review focuses on the role that the mitochondrial genome plays especially during early stages of development and in cancer.
Collapse
Affiliation(s)
- Justin C St John
- Centre for Genetic Diseases, Hudson Institute of Medical Research, and the Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, VIC 3168, Australia.
| |
Collapse
|
18
|
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: 0.8] [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
|
19
|
Wang HW, Xu Y, Miao YL, Luo HY, Wang KH. Mitochondrial DNA Haplogroup A may confer a genetic susceptibility to AIDS group from Southwest China. Mitochondrial DNA A DNA Mapp Seq Anal 2014; 27:2221-4. [PMID: 25431816 DOI: 10.3109/19401736.2014.982630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The acquired immunodeficiency syndrome (AIDS) in humans was one of the chronic infections caused by human immunodeficiency virus (HIV), and the interactions between viral infection and mitochondrial energetic implicated that mitochondrial DNA (mtDNA) variation(s) may effect genetic susceptibility to AIDS. Thus, to illustrate the maternal genetic structure and further identify whether mtDNA variation(s) can effect HIV infection among southwest Chinese AIDS group, the whole mtDNA control region sequences of 70 AIDS patients and 480 health individuals from southwest China were analyzed here. Our results indicated the plausible recent genetic admixture results of AIDS group; comparison of matrilineal components between AIDS and matched Han groups showed that mtDNA haplogroup A (p = 0.048, OR = 3.006, 95% CI = 1.109-8.145) has a significant higher difference between the two groups; further comparison illustrated that mtDNA mutations 16,209 (p = 0.046, OR = 2.607, 95% CI = 0.988-6.876) and 16,319 (p = 0.009, OR = 2.965, 95% CI = 1.278-6.876) have significant differences between AIDS and matched control groups, and both of which were the defining variations of mtDNA haplogroup A, they further confirmed that mtDNA haplogroup A may confer genetic susceptibility to AIDS. Our results suggested that haplogroup A may confer a genetic susceptibility to AIDS group from Southwest China.
Collapse
Affiliation(s)
- Hua-Wei Wang
- a Yunnan Institute of Digestive Disease, the First Affiliated Hospital of Kunming Medical University , Yunnan Province , China
| | - Yu Xu
- a Yunnan Institute of Digestive Disease, the First Affiliated Hospital of Kunming Medical University , Yunnan Province , China
| | - Ying-Lei Miao
- a Yunnan Institute of Digestive Disease, the First Affiliated Hospital of Kunming Medical University , Yunnan Province , China
| | - Hua-You Luo
- a Yunnan Institute of Digestive Disease, the First Affiliated Hospital of Kunming Medical University , Yunnan Province , China
| | - Kun-Hua Wang
- a Yunnan Institute of Digestive Disease, the First Affiliated Hospital of Kunming Medical University , Yunnan Province , China
| |
Collapse
|
20
|
Christiansen M, Hagen CM, Hedley PL. Mitochondrial haplogroups are associated with hypertrophic cardiomyopathy in the Indian population. Mitochondrion 2014; 20:105-6. [PMID: 25110059 DOI: 10.1016/j.mito.2014.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/30/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Michael Christiansen
- Department of Clinical Biochemistry, Immunology and Genetics (KBIG), Statens Serum Institut, Copenhagen, Denmark.
| | - Christian M Hagen
- Department of Clinical Biochemistry, Immunology and Genetics (KBIG), Statens Serum Institut, Copenhagen, Denmark
| | - Paula L Hedley
- Department of Clinical Biochemistry, Immunology and Genetics (KBIG), Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
21
|
Kenney MC, Chwa M, Atilano SR, Falatoonzadeh P, Ramirez C, Malik D, Tarek M, Cáceres-del-Carpio J, Nesburn AB, Boyer DS, Kuppermann BD, Vawter M, Jazwinski SM, Miceli M, Wallace DC, Udar N. Inherited mitochondrial DNA variants can affect complement, inflammation and apoptosis pathways: insights into mitochondrial-nuclear interactions. Hum Mol Genet 2014; 23:3537-51. [PMID: 24584571 PMCID: PMC4049308 DOI: 10.1093/hmg/ddu065] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/03/2014] [Accepted: 02/10/2014] [Indexed: 12/21/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss in developed countries. While linked to genetic polymorphisms in the complement pathway, there are many individuals with high risk alleles that do not develop AMD, suggesting that other 'modifiers' may be involved. Mitochondrial (mt) haplogroups, defined by accumulations of specific mtDNA single nucleotide polymorphisms (SNPs) which represent population origins, may be one such modifier. J haplogroup has been associated with high risk for AMD while the H haplogroup is protective. It has been difficult to assign biological consequences for haplogroups so we created human ARPE-19 cybrids (cytoplasmic hybrids), which have identical nuclei but mitochondria of either J or H haplogroups, to investigate their effects upon bioenergetics and molecular pathways. J cybrids have altered bioenergetic profiles compared with H cybrids. Q-PCR analyses show significantly lower expression levels for seven respiratory complex genes encoded by mtDNA. J and H cybrids have significantly altered expression of eight nuclear genes of the alternative complement, inflammation and apoptosis pathways. Sequencing of the entire mtDNA was carried out for all the cybrids to identify haplogroup and non-haplogroup defining SNPs. mtDNA can mediate cellular bioenergetics and expression levels of nuclear genes related to complement, inflammation and apoptosis. Sequencing data suggest that observed effects are not due to rare mtDNA variants but rather the combination of SNPs representing the J versus H haplogroups. These findings represent a paradigm shift in our concepts of mt-nuclear interactions.
Collapse
Affiliation(s)
- M Cristina Kenney
- Gavin Herbert Eye Institute, Department of Pathology and Laboratory Medicine,
| | | | | | | | | | | | | | | | - Anthony B Nesburn
- Gavin Herbert Eye Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David S Boyer
- Retina-Vitreous Associates Medical Group, Beverly Hills, CA, USA
| | | | - Marquis Vawter
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
| | | | - Michael Miceli
- Tulane Center for Aging, Tulane University, New Orleans, LA, USA
| | - Douglas C Wallace
- Children's Hospital of Philadelphia, Center for Mitochondrial and Epigenomic Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
22
|
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: 115] [Impact Index Per Article: 10.5] [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
|
23
|
Horan MP, Cooper DN. The emergence of the mitochondrial genome as a partial regulator of nuclear function is providing new insights into the genetic mechanisms underlying age-related complex disease. Hum Genet 2013; 133:435-58. [DOI: 10.1007/s00439-013-1402-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/23/2013] [Indexed: 12/17/2022]
|
24
|
Govindaraj P, Khan NA, Rani B, Rani DS, Selvaraj P, Jyothi V, Bahl A, Narasimhan C, Rakshak D, Premkumar K, Khullar M, Thangaraj K. Mitochondrial DNA variations associated with hypertrophic cardiomyopathy. Mitochondrion 2013; 16:65-72. [PMID: 24215792 DOI: 10.1016/j.mito.2013.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 10/17/2013] [Accepted: 10/29/2013] [Indexed: 01/11/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary disorder, characterized by unexplained hypertrophy of the left ventricle that frequently involved in the inter-ventricular septum. Mitochondrial DNA (mtDNA) mutations and haplogroups have been found to be associated with several diseases. Therefore, in the present study, we have sequenced the complete mtDNA of 114 clinically well-characterized HCM patients to look for the role of mtDNA variations and haplogroups in HCM phenotype among Indian patients. Complete mtDNA analysis revealed 28 novel variations, 25 disease-associated and 50 private mutations. We found 13 (11.40%) HCM patients having novel non-synonymous and/or MT-tRNA variations, of which two (m.4797C>M and m.8728T>Y) were in heteroplasmic condition. In silico prediction showed that a few mutations are pathogenic, which may affect the energy production in the heart. Unlike some of the other studies, we did not find association of mitochondrial haplogroup with HCM.
Collapse
Affiliation(s)
- Periyasamy Govindaraj
- Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, India
| | | | - Bindu Rani
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Deepa Selvi Rani
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | | - Ajay Bahl
- Department of Cardiology, PGIMER, Chandigarh, India
| | | | | | - Kumpati Premkumar
- Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | |
Collapse
|