1
|
Hong YS, Pasca S, Shi W, Puiu D, Lake NJ, Lek M, Ru M, Grove ML, Prizment A, Joshu CE, Platz EA, Guallar E, Arking DE, Gondek LP. Mitochondrial heteroplasmy improves risk prediction for myeloid neoplasms. Nat Commun 2024; 15:10133. [PMID: 39578475 PMCID: PMC11584845 DOI: 10.1038/s41467-024-54443-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Clonal hematopoiesis of indeterminate potential is the primary pathogenic risk factor for myeloid neoplasms, while heteroplasmy (mutations in a subset of cellular mitochondrial DNA) is another marker of clonal expansion associated with hematological malignancies. We explore how these two markers relate and influence myeloid neoplasms incidence, and their role in risk stratification. We find that heteroplasmy is more common in individuals with clonal hematopoiesis of indeterminate potential, particularly those with higher variant allele fractions, multiple mutations, or spliceosome machinery mutations. Individuals with both markers have a higher risk of myeloid neoplasms than those with either alone. Furthermore, heteroplasmic variants with higher predicted deleteriousness increase the risk of myeloid neoplasms. Incorporating heteroplasmy in an existing risk score model for individuals with clonal hematopoiesis of indeterminate potential significantly improves sensitivity and better identifies high-risk groups. This suggests heteroplasmy as a clonal expansion marker and potentially as a biomarker for myeloid neoplasms development.
Collapse
Affiliation(s)
- Yun Soo Hong
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sergiu Pasca
- Division of Hematological Malignancies, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wen Shi
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniela Puiu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Nicole J Lake
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Meng Ru
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Megan L Grove
- Human Genetics Center; Department of Epidemiology, Human Genetics, and Environmental Sciences; School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anna Prizment
- Department of Laboratory Medicine & Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Corinne E Joshu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth A Platz
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Eliseo Guallar
- Department of Epidemiology, School of Global Public Health, New York University, New York, NY, USA
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Lukasz P Gondek
- Division of Hematological Malignancies, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Jin X, Zhang R, Fu Y, Zhu Q, Hong L, Wu A, Wang H. Unveiling aging dynamics in the hematopoietic system insights from single-cell technologies. Brief Funct Genomics 2024; 23:639-650. [PMID: 38688725 DOI: 10.1093/bfgp/elae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
As the demographic structure shifts towards an aging society, strategies aimed at slowing down or reversing the aging process become increasingly essential. Aging is a major predisposing factor for many chronic diseases in humans. The hematopoietic system, comprising blood cells and their associated bone marrow microenvironment, intricately participates in hematopoiesis, coagulation, immune regulation and other physiological phenomena. The aging process triggers various alterations within the hematopoietic system, serving as a spectrum of risk factors for hematopoietic disorders, including clonal hematopoiesis, immune senescence, myeloproliferative neoplasms and leukemia. The emerging single-cell technologies provide novel insights into age-related changes in the hematopoietic system. In this review, we summarize recent studies dissecting hematopoietic system aging using single-cell technologies. We discuss cellular changes occurring during aging in the hematopoietic system at the levels of the genomics, transcriptomics, epigenomics, proteomics, metabolomics and spatial multi-omics. Finally, we contemplate the future prospects of single-cell technologies, emphasizing the impact they may bring to the field of hematopoietic system aging research.
Collapse
Affiliation(s)
- Xinrong Jin
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Ruohan Zhang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Yunqi Fu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Qiunan Zhu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Liquan Hong
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Aiwei Wu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Hu Wang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Deqing Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
3
|
Bertolini M, Clark D. Periodontal disease as a model to study chronic inflammation in aging. GeroScience 2024; 46:3695-3709. [PMID: 37285008 PMCID: PMC11226587 DOI: 10.1007/s11357-023-00835-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/20/2023] [Indexed: 06/08/2023] Open
Abstract
Periodontal disease is a chronic inflammatory condition that results in the destruction of the teeth supporting tissues, eventually leading to the loss of teeth and reduced quality of life. In severe cases, periodontal disease can limit proper nutritional intake, cause acute pain and infection, and cause a withdrawal from social situations due to esthetic and phonetic concerns. Similar to other chronic inflammatory conditions, periodontal disease increases in prevalence with age. Research into what drives periodontal disease pathogenesis in older adults is contributing to our general understanding of age-related chronic inflammation. This review will present periodontal disease as an age-related chronic inflammatory disease and as an effective geroscience model to study mechanisms of age-related inflammatory dysregulation. The current understanding of the cellular and molecular mechanisms that drive inflammatory dysregulation as a function of age will be discussed with a focus on the major pathogenic immune cells in periodontal disease, which include neutrophils, macrophages, and T cells. Research in the aging biology field has shown that the age-related changes in these immune cells result in the cells becoming less effective in the clearance of microbial pathogens, expansion of pathogenic subpopulations, or an increase in pro-inflammatory cytokine secretions. Such changes can be pathogenic and contribute to inflammatory dysregulation that is associated with a myriad of age-related disease including periodontal disease. An improved understanding is needed to develop better interventions that target the molecules or pathways that are perturbed with age in order to improve treatment of chronic inflammatory conditions, including periodontal disease, in older adult populations.
Collapse
Affiliation(s)
- Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Daniel Clark
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Nogueira C, Pereira C, Silva L, Laranjeira M, Lopes A, Neiva R, Rodrigues E, Campos T, Martins E, Bandeira A, Coelho M, Magalhães M, Damásio J, Gaspar A, Janeiro P, Gomes AL, Ferreira AC, Jacinto S, Vieira JP, Diogo L, Santos H, Mendonça C, Vilarinho L. The genetic landscape of mitochondrial diseases in the next-generation sequencing era: a Portuguese cohort study. Front Cell Dev Biol 2024; 12:1331351. [PMID: 38465286 PMCID: PMC10920333 DOI: 10.3389/fcell.2024.1331351] [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: 10/31/2023] [Accepted: 01/30/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction: Rare disorders that are genetically and clinically heterogeneous, such as mitochondrial diseases (MDs), have a challenging diagnosis. Nuclear genes codify most proteins involved in mitochondrial biogenesis, despite all mitochondria having their own DNA. The development of next-generation sequencing (NGS) technologies has revolutionized the understanding of many genes involved in the pathogenesis of MDs. In this new genetic era, using the NGS approach, we aimed to identify the genetic etiology for a suspected MD in a cohort of 450 Portuguese patients. Methods: We examined 450 patients using a combined NGS strategy, starting with the analysis of a targeted mitochondrial panel of 213 nuclear genes, and then proceeding to analyze the whole mitochondrial DNA. Results and Discussion: In this study, we identified disease-related variants in 134 (30%) analyzed patients, 88 with nuclear DNA (nDNA) and 46 with mitochondrial DNA (mtDNA) variants, most of them being pediatric patients (66%), of which 77% were identified in nDNA and 23% in mtDNA. The molecular analysis of this cohort revealed 72 already described pathogenic and 20 novel, probably pathogenic, variants, as well as 62 variants of unknown significance. For this cohort of patients with suspected MDs, the use of a customized gene panel provided a molecular diagnosis in a timely and cost-effective manner. Patients who cannot be diagnosed after this initial approach will be further selected for whole-exome sequencing. Conclusion: As a national laboratory for the study and research of MDs, we demonstrated the power of NGS to achieve a molecular etiology, expanding the mutational spectrum and proposing accurate genetic counseling in this group of heterogeneous diseases without therapeutic options.
Collapse
Affiliation(s)
- C. Nogueira
- Research & Development Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - C. Pereira
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - L. Silva
- Research & Development Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - Mateus Laranjeira
- Research & Development Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - A. Lopes
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - R. Neiva
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - E. Rodrigues
- Inherited Metabolic Diseases Reference Centre, São João Hospital University Centre, Porto, Portugal
| | - T. Campos
- Inherited Metabolic Diseases Reference Centre, São João Hospital University Centre, Porto, Portugal
| | - E. Martins
- Inherited Metabolic Diseases Reference Centre, Santo António Hospital University Centre, Porto, Portugal
| | - A. Bandeira
- Inherited Metabolic Diseases Reference Centre, Santo António Hospital University Centre, Porto, Portugal
| | - M. Coelho
- Inherited Metabolic Diseases Reference Centre, Santo António Hospital University Centre, Porto, Portugal
| | - M. Magalhães
- Neurology Department, Santo António Hospital University Centre, Porto, Portugal
| | - J. Damásio
- Neurology Department, Santo António Hospital University Centre, Porto, Portugal
| | - A. Gaspar
- Inherited Metabolic Diseases Reference Centre, Lisboa Norte Hospital University Centre, Lisboa, Portugal
| | - P Janeiro
- Inherited Metabolic Diseases Reference Centre, Lisboa Norte Hospital University Centre, Lisboa, Portugal
| | - A. Levy Gomes
- Neurology Department, Lisboa Norte Hospital University Centre, Lisboa, Portugal
| | - A. C. Ferreira
- Inherited Metabolic Diseases Reference Centre, Lisboa Central Hospital Centre, Lisboa, Portugal
| | - S. Jacinto
- Inherited Metabolic Diseases Reference Centre, Lisboa Central Hospital Centre, Lisboa, Portugal
| | - J. P. Vieira
- Inherited Metabolic Diseases Reference Centre, Lisboa Central Hospital Centre, Lisboa, Portugal
| | - L. Diogo
- Inherited Metabolic Diseases Reference Centre, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - H. Santos
- Inherited Metabolic Diseases Reference Centre, Vila Nova de Gaia Hospital Centre, Vila Nova de Gaia, Portugal
| | - C. Mendonça
- Pediatric Department, Faro Hospital and University Centre, Faro, Portugal
| | - L. Vilarinho
- Research & Development Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| |
Collapse
|
5
|
Hernández CL. Mitochondrial DNA in Human Diversity and Health: From the Golden Age to the Omics Era. Genes (Basel) 2023; 14:1534. [PMID: 37628587 PMCID: PMC10453943 DOI: 10.3390/genes14081534] [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: 06/19/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrial DNA (mtDNA) is a small fraction of our hereditary material. However, this molecule has had an overwhelming presence in scientific research for decades until the arrival of high-throughput studies. Several appealing properties justify the application of mtDNA to understand how human populations are-from a genetic perspective-and how individuals exhibit phenotypes of biomedical importance. Here, I review the basics of mitochondrial studies with a focus on the dawn of the field, analysis methods and the connection between two sides of mitochondrial genetics: anthropological and biomedical. The particularities of mtDNA, with respect to inheritance pattern, evolutionary rate and dependence on the nuclear genome, explain the challenges of associating mtDNA composition and diseases. Finally, I consider the relevance of this single locus in the context of omics research. The present work may serve as a tribute to a tool that has provided important insights into the past and present of humankind.
Collapse
Affiliation(s)
- Candela L Hernández
- Department of Biodiversity, Ecology and Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
6
|
Malyutina S, Maximov V, Chervova O, Orlov P, Ivanova A, Mazdorova E, Ryabikov A, Simonova G, Voevoda M. The Relationship between All-Cause Natural Mortality and Copy Number of Mitochondrial DNA in a 15-Year Follow-Up Study. Int J Mol Sci 2023; 24:10469. [PMID: 37445647 DOI: 10.3390/ijms241310469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
We explored the relationship between the copy number of mitochondrial DNA (mtDNA-CN) and all-cause natural mortality. We examined a random population sample in 2003/2005 (n = 9360, men/women, 45-69, the HAPIEE project) and followed up for 15 years. Using a nested case-control design, we selected non-external deaths among those free from baseline cardiovascular diseases (CVD) and cancer (n = 371), and a sex- and age-stratified control (n = 785). The odds ratios (ORs) of death were 1.06 (95%CI 1.01-1.11) per one-decile decrease in mtDNA-CN independent of age, sex, metabolic factors, smoking, alcohol intake and education. The age-sex-adjusted ORs of death in the second and first tertiles of mtDNA-CN vs. the top tertile were 2.35 (95% CI 1.70-3.26) and 1.59 (1.16-2.17); an increased risk was confined to the second tertile after controlling for smoking and metabolic factors. The multivariable-adjusted OR of CVD death was 1.92 (95% CI 1.18-3.15) in tertile 2 vs. the top tertile of mtDNA-CN, and for cancer-related death the ORs were 3.66 (95% CI 2.21-6.05) and 2.29 (95% CI 1.43-3.68) in tertiles 2 and 1 vs. the top tertile. In the Siberian population cohort, the mtDNA-CN was an inverse predictor of the 15-year risk of natural mortality, due to the greatest impact of CVD and cancer-related death. The findings merit attention for exploring further the role of mtDNA in human ageing and the diversity of mortality.
Collapse
Affiliation(s)
- Sofia Malyutina
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| | - Vladimir Maximov
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| | - Olga Chervova
- UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - Pavel Orlov
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| | - Anastasiya Ivanova
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| | - Ekaterina Mazdorova
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| | - Andrew Ryabikov
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| | - Galina Simonova
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| | - Mikhail Voevoda
- Research Institute of Internal and Preventive Medicine-Branch of Institute of Cytology and Genetics SB RAS, Novosibirsk 630089, Russia
| |
Collapse
|
7
|
San-Millán I. The Key Role of Mitochondrial Function in Health and Disease. Antioxidants (Basel) 2023; 12:antiox12040782. [PMID: 37107158 PMCID: PMC10135185 DOI: 10.3390/antiox12040782] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
The role of mitochondrial function in health and disease has become increasingly recognized, particularly in the last two decades. Mitochondrial dysfunction as well as disruptions of cellular bioenergetics have been shown to be ubiquitous in some of the most prevalent diseases in our society, such as type 2 diabetes, cardiovascular disease, metabolic syndrome, cancer, and Alzheimer's disease. However, the etiology and pathogenesis of mitochondrial dysfunction in multiple diseases have yet to be elucidated, making it one of the most significant medical challenges in our history. However, the rapid advances in our knowledge of cellular metabolism coupled with the novel understanding at the molecular and genetic levels show tremendous promise to one day elucidate the mysteries of this ancient organelle in order to treat it therapeutically when needed. Mitochondrial DNA mutations, infections, aging, and a lack of physical activity have been identified to be major players in mitochondrial dysfunction in multiple diseases. This review examines the complexities of mitochondrial function, whose ancient incorporation into eukaryotic cells for energy purposes was key for the survival and creation of new species. Among these complexities, the tightly intertwined bioenergetics derived from the combustion of alimentary substrates and oxygen are necessary for cellular homeostasis, including the production of reactive oxygen species. This review discusses different etiological mechanisms by which mitochondria could become dysregulated, determining the fate of multiple tissues and organs and being a protagonist in the pathogenesis of many non-communicable diseases. Finally, physical activity is a canonical evolutionary characteristic of humans that remains embedded in our genes. The normalization of a lack of physical activity in our modern society has led to the perception that exercise is an "intervention". However, physical activity remains the modus vivendi engrained in our genes and being sedentary has been the real intervention and collateral effect of modern societies. It is well known that a lack of physical activity leads to mitochondrial dysfunction and, hence, it probably becomes a major etiological factor of many non-communicable diseases affecting modern societies. Since physical activity remains the only stimulus we know that can improve and maintain mitochondrial function, a significant emphasis on exercise promotion should be imperative in order to prevent multiple diseases. Finally, in populations with chronic diseases where mitochondrial dysfunction is involved, an individualized exercise prescription should be crucial for the "metabolic rehabilitation" of many patients. From lessons learned from elite athletes (the perfect human machines), it is possible to translate and apply multiple concepts to the betterment of populations with chronic diseases.
Collapse
Affiliation(s)
- Iñigo San-Millán
- Department of Human Physiology and Nutrition, University of Colorado, Colorado Springs, CO 80198, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Long read mitochondrial genome sequencing using Cas9-guided adaptor ligation. Mitochondrion 2022; 65:176-183. [PMID: 35787470 DOI: 10.1016/j.mito.2022.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/11/2022] [Accepted: 06/19/2022] [Indexed: 11/21/2022]
Abstract
The mitochondrial genome (mtDNA) is an important source of disease-causing genetic variability, but existing sequencing methods limit understanding, precluding phased measurement of mutations and clear detection of large sporadic deletions. We adapted a method for amplification-free sequence enrichment using Cas9 cleavage to obtain full length nanopore reads of mtDNA. We then utilized the long reads to phase mutations in a patient with an mtDNA-linked syndrome and demonstrated that this method can map age-induced mtDNA deletions. We believe this method will offer deeper insight into our understanding of mtDNA variation.
Collapse
|
9
|
Abstract
Mitochondria are the main source of energy used to maintain cellular homeostasis. This aspect of mitochondrial biology underlies their putative role in age-associated tissue dysfunction. Proper functioning of the electron transport chain (ETC), which is partially encoded by the extra-nuclear mitochondrial genome (mtDNA), is key to maintaining this energy production. The acquisition of de novo somatic mutations that interrupt the function of the ETC have long been associated with aging and common diseases of the elderly. Yet, despite over 30 years of study, the exact role(s) mtDNA mutations play in driving aging and its associated pathologies remains under considerable debate. Furthermore, even fundamental aspects of age-related mtDNA mutagenesis, such as when mutations arise during aging, where and how often they occur across tissues, and the specific mechanisms that give rise to them, remain poorly understood. In this review, we address the current understanding of the somatic mtDNA mutations, with an emphasis of when, where, and how these mutations arise during aging. Additionally, we highlight current limitations in our knowledge and critically evaluate the controversies stemming from these limitations. Lastly, we highlight new and emerging technologies that offer potential ways forward in increasing our understanding of somatic mtDNA mutagenesis in the aging process.
Collapse
Affiliation(s)
- Monica Sanchez-Contreras
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Scott R Kennedy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| |
Collapse
|
10
|
Sanchez-Contreras M, Sweetwyne MT, Kohrn BF, Tsantilas KA, Hipp MJ, Schmidt EK, Fredrickson J, Whitson JA, Campbell MD, Rabinovitch PS, Marcinek DJ, Kennedy SR. A replication-linked mutational gradient drives somatic mutation accumulation and influences germline polymorphisms and genome composition in mitochondrial DNA. Nucleic Acids Res 2021; 49:11103-11118. [PMID: 34614167 PMCID: PMC8565317 DOI: 10.1093/nar/gkab901] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 11/22/2022] Open
Abstract
Mutations in mitochondrial DNA (mtDNA) cause maternally inherited diseases, while somatic mutations are linked to common diseases of aging. Although mtDNA mutations impact health, the processes that give rise to them are under considerable debate. To investigate the mechanism by which de novo mutations arise, we analyzed the distribution of naturally occurring somatic mutations across the mouse and human mtDNA obtained by Duplex Sequencing. We observe distinct mutational gradients in G→A and T→C transitions delimited by the light-strand origin and the mitochondrial Control Region (mCR). The gradient increases unequally across the mtDNA with age and is lost in the absence of DNA polymerase γ proofreading activity. In addition, high-resolution analysis of the mCR shows that important regulatory elements exhibit considerable variability in mutation frequency, consistent with them being mutational ‘hot-spots’ or ‘cold-spots’. Collectively, these patterns support genome replication via a deamination prone asymmetric strand-displacement mechanism as the fundamental driver of mutagenesis in mammalian DNA. Moreover, the distribution of mtDNA single nucleotide polymorphisms in humans and the distribution of bases in the mtDNA across vertebrate species mirror this gradient, indicating that replication-linked mutations are likely the primary source of inherited polymorphisms that, over evolutionary timescales, influences genome composition during speciation.
Collapse
Affiliation(s)
- Monica Sanchez-Contreras
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Mariya T Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Brendan F Kohrn
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Michael J Hipp
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth K Schmidt
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Jeanne Fredrickson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Jeremy A Whitson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Matthew D Campbell
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Peter S Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, WA 98195, USA
| | - Scott R Kennedy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
11
|
Van Remmen H, Freeman WM, Miller BF, Kinter M, Wren JD, Chiao A, Towner RA, Snider TA, Sonntag WE, Richardson A. Oklahoma Nathan Shock Aging Center - assessing the basic biology of aging from genetics to protein and function. GeroScience 2021; 43:2183-2203. [PMID: 34606039 PMCID: PMC8599778 DOI: 10.1007/s11357-021-00454-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 01/21/2023] Open
Abstract
The Oklahoma Shock Nathan Shock Center is designed to deliver unique, innovative services that are not currently available at most institutions. The focus of the Center is on geroscience and the development of careers of young investigators. Pilot grants are provided through the Research Development Core to junior investigators studying aging/geroscience throughout the USA. However, the services of our Center are available to the entire research community studying aging and geroscience. The Oklahoma Nathan Shock Center provides researchers with unique services through four research cores. The Multiplexing Protein Analysis Core uses the latest mass spectrometry technology to simultaneously measure the levels, synthesis, and turnover of hundreds of proteins associated with pathways of importance to aging, e.g., metabolism, antioxidant defense system, proteostasis, and mitochondria function. The Genomic Sciences Core uses novel next-generation sequencing that allows investigators to study the effect of age, or anti-aging manipulations, on DNA methylation, mitochondrial genome heteroplasmy, and the transcriptome of single cells. The Geroscience Redox Biology Core provides investigators with a comprehensive state-of-the-art assessment of the oxidative stress status of a cell, e.g., measures of oxidative damage and redox couples, which are important in aging as well as many major age-related diseases as well as assays of mitochondrial function. The GeroInformatics Core provides investigators assistance with data analysis, which includes both statistical support as well as analysis of large datasets. The Core also has developed number of unique software packages to help with interpretation of results and discovery of new leads relevant to aging. In addition, the Geropathology Research Resource in the Program Enhancement Core provides investigators with pathological assessments of mice using the recently developed Geropathology Grading Platform.
Collapse
Affiliation(s)
- Holly Van Remmen
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
- Biochemistry & Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma, City, OK, USA.
| | - Willard M Freeman
- Biochemistry & Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma, City, OK, USA
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Benjamin F Miller
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Biochemistry & Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma, City, OK, USA
| | - Michael Kinter
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Jonathan D Wren
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ann Chiao
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Timothy A Snider
- Department of Veterinary Pathology, Center for Veterinary Health Sciences at, Oklahoma State University, Stillwater, OK, USA
| | - William E Sonntag
- Biochemistry & Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma, City, OK, USA
| | - Arlan Richardson
- Biochemistry & Molecular Biology, The University of Oklahoma Health Sciences Center, Oklahoma, City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
12
|
Lagunas-Rangel FA. Deciphering the whale's secrets to have a long life. Exp Gerontol 2021; 151:111425. [PMID: 34051285 DOI: 10.1016/j.exger.2021.111425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 01/20/2023]
Abstract
Whales are marine creatures known for their enormous size and that live in all the oceans on earth. One of the oldest known organisms is bowhead whales, which can survive up to 200 years, and similarly, other species of whales have shown a remarkable long lifespan. In addition to this, whales are highly resistant to cancer, a disease that is strongly related to aging and the accumulation of damage over time. These two characteristics make whales an interesting model to study and that can provide us with a track both to delay aging and to avoid pathologies associated with it, such as cancer. In the present work, we try to analyze different aspects of whales such as metabolism, hematological and biochemical characteristics, and properties of their genome and transcriptome in order to elucidate possible molecular mechanisms that evolution has provided to these aquatic mammals.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico; Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
13
|
Brandts L, van Poppel FW, van den Brandt PA. Parental lifespan and the likelihood of reaching the age of 90 years in the Netherlands Cohort Study. Geriatr Gerontol Int 2020; 21:215-221. [PMID: 33368897 PMCID: PMC7898670 DOI: 10.1111/ggi.14120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 10/12/2020] [Accepted: 11/29/2020] [Indexed: 12/16/2022]
Abstract
Aim Growing evidence suggests an association between parental longevity and lifespan of subsequent generations. We aimed to reproduce earlier findings, showing a positive association between parental longevity and offspring's longevity. Additionally, we investigated whether this is mainly driven by the maternal or paternal germline in male and female offspring. Methods For these analyses, data from the oldest birth cohort (1916–17) of the Netherlands Cohort Study was used. Participants filled in a baseline questionnaire in 1986 (at age 68–70 years). Follow up for vital status information until the age of 90 years (2006–07) was >99.9% complete. Multivariable‐adjusted Cox regression analyses with a fixed follow‐up time were based on 2368 men and 2657 women with complete parental survival data and relevant confounders to calculate risk ratios (RR) of reaching longevity. Results In age‐adjusted models, paternal and maternal age at death were significantly positively associated with reaching 90 years in both male and female offspring. In male offspring, paternal age at death (≥90 years vs <80 years) showed the strongest association with survival to 90 years (RR 1.42, 95% CI 1.07–1.89), after confounder correction. In female offspring, maternal age at death (≥90 years vs <80 years) showed the strongest association with survival to 90 years (RR 1.20, 95% CI 1.04–1.40). Discussion After confounder adjustment, stronger and significant associations were observed between paternal lifespan and male offspring longevity, and maternal lifespan and female offspring longevity. Future research should investigate through which pathways a longer lifespan of parents is transmitted to their offspring. Geriatr Gerontol Int 2021; 21: 215–221.
Collapse
Affiliation(s)
- Lloyd Brandts
- GROW - School for Oncology and Developmental Biology, Department of Epidemiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Frans Wa van Poppel
- Netherlands Interdisciplinary Demographic Institute (NIDI)/Royal Netherlands Academy of Arts and Sciences (KNAW), The Hague, the Netherlands
| | - Piet A van den Brandt
- GROW - School for Oncology and Developmental Biology, Department of Epidemiology, Maastricht University Medical Center, Maastricht, the Netherlands.,CAPHRI - School for Public Health and Primary Care, Department of Epidemiology, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
14
|
Sebastian-Valverde M, Pasinetti GM. The NLRP3 Inflammasome as a Critical Actor in the Inflammaging Process. Cells 2020; 9:cells9061552. [PMID: 32604771 PMCID: PMC7348816 DOI: 10.3390/cells9061552] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/12/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
As a consequence of the considerable increase in the human lifespan over the last century, we are experiencing the appearance and impact of new age-related diseases. The causal relationships between aging and an enhanced susceptibility of suffering from a broad spectrum of diseases need to be better understood. However, one specific shared feature seems to be of capital relevance for most of these conditions: the low-grade chronic inflammatory state inherently associated with aging, i.e., inflammaging. Here, we review the molecular and cellular mechanisms that link aging and inflammaging, focusing on the role of the innate immunity and more concretely on the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, as well as how the chronic activation of this inflammasome has a detrimental effect on different age-related disorders.
Collapse
Affiliation(s)
| | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY 10468, USA
- Correspondence: ; Tel.: +1-212-241-1952
| |
Collapse
|
15
|
Autologous reference types can confound the detection of somatic mutation in solid cancers. DNA Repair (Amst) 2018; 69:6-13. [PMID: 30029072 DOI: 10.1016/j.dnarep.2018.07.002] [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: 05/25/2018] [Revised: 06/17/2018] [Accepted: 07/03/2018] [Indexed: 11/22/2022]
Abstract
Vast number of somatic mutations has been proved to be affected by the factors of sequencing methods, analysis pipelines and validation methods. We here showed the effect of autologous reference types on the detection of cancer-associated somatic mutations with the somatic single nucleotide variations (SNVs) and clinical data of solid tumors from the Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC). The distribution of somatic SNVs was significantly different among groups of autologous references in 6 cancers detected by whole genome sequencing (WGS) and 5 cancers detected by the random sequencing of exonic regions selected from the genome (WXS), especially in protein coding region of 5 cancers with age, gender and TNM adjusted. In addition, only 60.24% (95% CI: 49.65%-70.83%) of the somatic SNVs called from normal blood by WXS were found in those called from normal solid tissue tested by WXS / WGS, while 31.78% (95%CI: 4.14%-59.42%) of the somatic SNVs called from normal tissue adjacent to primary by WXS were found in those from normal blood tested by WXS / WGS. These findings suggested that more representative types of normal tissues should be included in detection of cancer-associated somatic mutations.
Collapse
|
16
|
Rueda M, Torkamani A. SG-ADVISER mtDNA: a web server for mitochondrial DNA annotation with data from 200 samples of a healthy aging cohort. BMC Bioinformatics 2017; 18:373. [PMID: 28821228 PMCID: PMC5563004 DOI: 10.1186/s12859-017-1778-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/31/2017] [Indexed: 02/03/2023] Open
Abstract
Background Whole genome and exome sequencing usually include reads containing mitochondrial DNA (mtDNA). Yet, state-of-the-art pipelines and services for human nuclear genome variant calling and annotation do not handle mitochondrial genome data appropriately. As a consequence, any researcher desiring to add mtDNA variant analysis to their investigations is forced to explore the literature for mtDNA pipelines, evaluate them, and implement their own instance of the desired tool. This task is far from trivial, and can be prohibitive for non-bioinformaticians. Results We have developed SG-ADVISER mtDNA, a web server to facilitate the analysis and interpretation of mtDNA genomic data coming from next generation sequencing (NGS) experiments. The server was built in the context of our SG-ADVISER framework and on top of the MtoolBox platform (Calabrese et al., Bioinformatics 30(21):3115–3117, 2014), and includes most of its functionalities (i.e., assembly of mitochondrial genomes, heteroplasmic fractions, haplogroup assignment, functional and prioritization analysis of mitochondrial variants) as well as a back-end and a front-end interface. The server has been tested with unpublished data from 200 individuals of a healthy aging cohort (Erikson et al., Cell 165(4):1002–1011, 2016) and their data is made publicly available here along with a preliminary analysis of the variants. We observed that individuals over ~90 years old carried low levels of heteroplasmic variants in their genomes. Conclusions SG-ADVISER mtDNA is a fast and functional tool that allows for variant calling and annotation of human mtDNA data coming from NGS experiments. The server was built with simplicity in mind, and builds on our own experience in interpreting mtDNA variants in the context of sudden death and rare diseases. Our objective is to provide an interface for non-bioinformaticians aiming to acquire (or contrast) mtDNA annotations via MToolBox. SG-ADVISER web server is freely available to all users at https://genomics.scripps.edu/mtdna. Electronic supplementary material The online version of this article (doi:10.1186/s12859-017-1778-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manuel Rueda
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - Ali Torkamani
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
17
|
Li H, Shen L, Hu P, Huang R, Cao Y, Deng J, Yuan W, Liu D, Yang J, Gu H, Bai Y. Aging-associated mitochondrial DNA mutations alter oxidative phosphorylation machinery and cause mitochondrial dysfunctions. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2266-2273. [PMID: 28559044 DOI: 10.1016/j.bbadis.2017.05.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Our previous study generated a series of cybrids containing mitochondria of synaptosomes from mice at different ages. The following functional analysis on these cybrids revealed an age-dependent decline of mitochondrial function. To understand the underlying mechanisms that contribute to the age-related mitochondrial dysfunction, we focused on three cybrids carrying mitochondria derived from synaptosomes of the old mice that exhibited severe respiratory deficiencies. In particular, we started with a comprehensive analysis of mitochondrial genome by high resolution, high sensitive deep sequencing method. Compared with young control, we detected a significant accumulation of heteroplasmic mtDNA mutations. These mutations included six alterations in main control region that has been shown to regulate overall gene-expression, and four alterations in protein coding region, two of which led to significant changes in complex I subunit ND5 and complex III subunit CytB. Interestingly, a reduced mtDNA-encoded protein synthesis was associated with the changes in the main control region. Likewise, mutations in ND5 and CytB were associated with defects in assembly of respiratory complexes. Altogether, the identified age-dependent accumulation of mtDNA mutations in mouse brain likely contributes to the decline in mitochondrial function.
Collapse
Affiliation(s)
- Hongzhi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Luxi Shen
- Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Peiqing Hu
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Rong Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ying Cao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Janice Deng
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Weihua Yuan
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Danhui Liu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jifeng Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Haihua Gu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yidong Bai
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
18
|
Zinovkin RA, Skulachev MV, Skulachev VP. Mitochondrial Genome and Longevity. BIOCHEMISTRY (MOSCOW) 2017; 81:1401-1405. [PMID: 28259117 DOI: 10.1134/s0006297916120014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The mitochondrial genome provides not only respiratory chain function, but it also ensures the impact of mitochondria on nearly all crucial metabolic processes. It is well known that mitochondria regulate aging and lifespan. However, until now there were no direct experimental data concerning the influence of various mitochondrial DNA variants on lifespan of animals with identical nuclear genome. In a recent paper of J. A. Enríquez and coworkers (Latorre-Pellicer, A., et al. (2016) Nature, 535, 561-565), it was shown that mice carrying nuclear DNA from one strain and mitochondrial DNA from another had longer median lifespan and retarded development of various aging traits. This review critically analyzes that paper and considers some aspects of the crosstalk between the nuclear and mitochondrial genomes. We also discuss new perspectives of gerontology in the light of the discovery made by Enríquez's group.
Collapse
Affiliation(s)
- R A Zinovkin
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, 119991, Russia.
| | | | | |
Collapse
|
19
|
Bertolo A, Capossela S, Fränkl G, Baur M, Pötzel T, Stoyanov J. Oxidative status predicts quality in human mesenchymal stem cells. Stem Cell Res Ther 2017; 8:3. [PMID: 28061861 PMCID: PMC5217567 DOI: 10.1186/s13287-016-0452-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/24/2016] [Accepted: 12/06/2016] [Indexed: 01/20/2023] Open
Abstract
Background Human bone marrow-derived mesenchymal stem cells (MSC) are adult progenitor cells with great potential for application in cell-based therapies. From a cell-based therapy perspective, there are two limitations to MSC use: (1) these therapies require large numbers of cells, and long-term expansion of MSC in vitro promotes replicative senescence; and (2) patient variability is a challenge for defining MSC quality standards for transplantation. This study aimed to determine whether low or high oxidative status of MSC correlate with changes in cell expansion and differentiation potentials. Methods We investigated functional aspects of mitochondria, such as cell metabolic activity indicators and expression of antioxidant enzymes. Furthermore, we tested if senescence-induced changes in oxidative status of MSC could be counteracted by methylene blue (MB), an alternative mitochondrial electron transfer known to enhance cell bioenergetics. Results MSC isolated from donors of the same age showed distinctive behavior in culture and were grouped as weak (low colony-forming units (CFU) and a short life in vitro) and vigorous MSC (high CFU and a long life in vitro). In comparison to weak MSC, vigorous MSC had oxidative status characterized by lower mitochondrial membrane potential, lower mitochondrial activity, and fewer reactive oxygen species production, as well as reduced mitochondrial biogenesis. Vigorous MSC had a significantly higher expansion potential compared to weak MSC, while no differences were observed during differentiation. MB treatment significantly improved expansion and differentiation potential, however only in vigorous MSC. Conclusions Together, these results demonstrate the importance of mitochondrial function in MSC in vitro, and that cells with low oxidative status levels are better candidates for cell-based therapies.
Collapse
Affiliation(s)
- Alessandro Bertolo
- Biomedical laboratories, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Simona Capossela
- Biomedical laboratories, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Gion Fränkl
- Biomedical laboratories, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Martin Baur
- Cantonal Hospital of Lucerne, Lucerne, Switzerland.,Swiss Paraplegic Centre, Nottwil, Switzerland
| | | | - Jivko Stoyanov
- Biomedical laboratories, Swiss Paraplegic Research, Nottwil, Switzerland. .,Institute for Surgical Technology and Biomechanics, University of Bern, Bern, Switzerland.
| |
Collapse
|
20
|
Hefti E, Bard J, Blanco JG. Analysis of Heteroplasmic Variants in the Cardiac Mitochondrial Genome of Individuals with Down Syndrome. Hum Mutat 2016; 38:48-54. [PMID: 27594409 DOI: 10.1002/humu.23071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/29/2016] [Indexed: 11/08/2022]
Abstract
Individuals with Down syndrome (DS, trisomy 21) exhibit a pro-oxidative cellular environment as well as mitochondrial dysfunction. Increased oxidative stress may damage the mitochondrial DNA (mtDNA). The coexistence of mtDNA variants in a cell or tissue (i.e., heteroplasmy) may contribute to mitochondrial dysfunction. Given the evidence on mitochondrial dysfunction and the relatively high incidence of multiorganic disorders associated with DS, we hypothesized that cardiac tissue from subjects with DS may exhibit higher frequencies of mtDNA variants in comparison to cardiac tissue from donors without DS. This study documents the analysis of mtDNA variants in heart tissue samples from donors with (n = 12) and without DS (n = 33) using massively parallel sequencing. Contrary to the original hypothesis, the study's findings suggest that the cardiac mitochondrial genomes from individuals with and without DS exhibit many similarities in terms of (1) total number of mtDNA variants per sample, (2) the frequency of mtDNA variants, (3) the type of mtDNA variants, and (4) the patterns of distribution of mtDNA variants. In both groups of samples, the mtDNA control region showed significantly more heteroplasmic variants in comparison to the number of variants in protein- and RNA-coding genes (P < 1.00×10-4 , ANOVA).
Collapse
Affiliation(s)
- Erik Hefti
- Department of Pharmaceutical Sciences, The School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| | - Jonathan Bard
- University at Buffalo Genomics and Bioinformatics Core, New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York
| | - Javier G Blanco
- Department of Pharmaceutical Sciences, The School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
21
|
Zhang P, Samuels DC, Lehmann B, Stricker T, Pietenpol J, Shyr Y, Guo Y. Mitochondria sequence mapping strategies and practicability of mitochondria variant detection from exome and RNA sequencing data. Brief Bioinform 2016; 17:224-32. [PMID: 26249222 PMCID: PMC5974936 DOI: 10.1093/bib/bbv057] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/23/2015] [Indexed: 12/30/2022] Open
Abstract
The rapid progress in high-throughput sequencing has significantly enriched our capacity for studying the mitochondrial DNA (mtDNA). In addition to performing specific mitochondrial targeted sequencing, an increasingly popular alternative approach is using the off-target reads from exome sequencing to infer mtDNA variants, including single nucleotide polymorphisms (SNPs) and heteroplasmy. However, the effectiveness and practicality of this approach have not been tested. Recently, RNAseq data have also been suggested as a good source for alternative data mining, but whether mitochondrial variants can be detected from RNAseq data has not been validated. We designed a study to evaluate the practicability of mtDNA variant detection using exome and RNA sequencing data. Five breast cancer cell lines were sequenced through mitochondrial targeted, exome, and RNA sequencing. Mitochondrial targeted sequencing was used as the gold standard to compute the validation and false discovery rates of SNP and heteroplasmy detection in exome and RNAseq data. We found that exome and RNA sequencing can accurately detect mitochondrial SNPs. However, the lower false discovery rate makes exome sequencing a better choice for heteroplasmy detection than RNAseq. Furthermore, we examined three alignment strategies and found that aligning reads directly to the mitochondrial reference genome or aligning reads to the nuclear and mitochondrial references genomes simultaneously produced the best results, and that aligning to the nuclear genome first and afterwards to the mitochondrial genome performed poorly. In conclusion, our study provides important guidelines for future studies that intend to use either exome sequencing or RNAseq data to infer mitochondrial SNPs and heteroplasmy.
Collapse
|
22
|
Zhang P, Samuels DC, Lehmann B, Stricker T, Pietenpol J, Shyr Y, Guo Y. Practicality of identifying mitochondria variants from exome and RNAseq data. BMC Bioinformatics 2015. [PMCID: PMC4625201 DOI: 10.1186/1471-2105-16-s15-p6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
23
|
Toren D, Barzilay T, Tacutu R, Lehmann G, Muradian KK, Fraifeld VE. MitoAge: a database for comparative analysis of mitochondrial DNA, with a special focus on animal longevity. Nucleic Acids Res 2015; 44:D1262-5. [PMID: 26590258 PMCID: PMC4702847 DOI: 10.1093/nar/gkv1187] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/23/2015] [Indexed: 11/22/2022] Open
Abstract
Mitochondria are the only organelles in the animal cells that have their own genome. Due to a key role in energy production, generation of damaging factors (ROS, heat), and apoptosis, mitochondria and mtDNA in particular have long been considered one of the major players in the mechanisms of aging, longevity and age-related diseases. The rapidly increasing number of species with fully sequenced mtDNA, together with accumulated data on longevity records, provides a new fascinating basis for comparative analysis of the links between mtDNA features and animal longevity. To facilitate such analyses and to support the scientific community in carrying these out, we developed the MitoAge database containing calculated mtDNA compositional features of the entire mitochondrial genome, mtDNA coding (tRNA, rRNA, protein-coding genes) and non-coding (D-loop) regions, and codon usage/amino acids frequency for each protein-coding gene. MitoAge includes 922 species with fully sequenced mtDNA and maximum lifespan records. The database is available through the MitoAge website (www.mitoage.org or www.mitoage.info), which provides the necessary tools for searching, browsing, comparing and downloading the data sets of interest for selected taxonomic groups across the Kingdom Animalia. The MitoAge website assists in statistical analysis of different features of the mtDNA and their correlative links to longevity.
Collapse
Affiliation(s)
- Dmitri Toren
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Thomer Barzilay
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Robi Tacutu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Gilad Lehmann
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israel Tumor and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | | | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
24
|
Tower J. Programmed cell death in aging. Ageing Res Rev 2015; 23:90-100. [PMID: 25862945 DOI: 10.1016/j.arr.2015.04.002] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/15/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023]
Abstract
Programmed cell death (PCD) pathways, including apoptosis and regulated necrosis, are required for normal cell turnover and tissue homeostasis. Mis-regulation of PCD is increasingly implicated in aging and aging-related disease. During aging the cell turnover rate declines for several highly-mitotic tissues. Aging-associated disruptions in systemic and inter-cell signaling combined with cell-autonomous damage and mitochondrial malfunction result in increased PCD in some cell types, and decreased PCD in other cell types. Increased PCD during aging is implicated in immune system decline, skeletal muscle wasting (sarcopenia), loss of cells in the heart, and neurodegenerative disease. In contrast, cancer cells and senescent cells are resistant to PCD, enabling them to increase in abundance during aging. PCD pathways limit life span in fungi, but whether PCD pathways normally limit adult metazoan life span is not yet clear. PCD is regulated by a balance of negative and positive factors, including the mitochondria, which are particularly subject to aging-associated malfunction.
Collapse
|
25
|
Abstract
In the past century, considerable efforts were made to understand the role of mitochondrial DNA (mtDNA) mutations and of oxidative stress in aging. The classic mitochondrial free radical theory of aging, in which mtDNA mutations cause genotoxic oxidative stress, which in turn creates more mutations, has been a central hypothesis in the field for decades. In the past few years, however, new elements have discredited this original theory. The major sources of mitochondrial DNA mutations seem to be replication errors and failure of the repair mechanisms, and the accumulation of these mutations as observed in aged organisms seems to occur by clonal expansion and not to be caused by a reactive oxygen species-dependent vicious cycle. New hypotheses of how age-associated mitochondrial dysfunction may lead to aging are based on the role of reactive oxygen species as signaling molecules and on their role in mediating stress responses to age-dependent damage. Here, we review the changes that mtDNA undergoes during aging and the past and most recent hypotheses linking these changes to the tissue failure observed in aging.
Collapse
Affiliation(s)
- Milena Pinto
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Carlos T Moraes
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Department of Cell Biology and Anatomy, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
26
|
Mitochondrial Oxidative Stress, Mitochondrial DNA Damage and Their Role in Age-Related Vascular Dysfunction. Int J Mol Sci 2015; 16:15918-53. [PMID: 26184181 PMCID: PMC4519931 DOI: 10.3390/ijms160715918] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/17/2015] [Accepted: 06/29/2015] [Indexed: 02/06/2023] Open
Abstract
The prevalence of cardiovascular diseases is significantly increased in the older population. Risk factors and predictors of future cardiovascular events such as hypertension, atherosclerosis, or diabetes are observed with higher frequency in elderly individuals. A major determinant of vascular aging is endothelial dysfunction, characterized by impaired endothelium-dependent signaling processes. Increased production of reactive oxygen species (ROS) leads to oxidative stress, loss of nitric oxide (•NO) signaling, loss of endothelial barrier function and infiltration of leukocytes to the vascular wall, explaining the low-grade inflammation characteristic for the aged vasculature. We here discuss the importance of different sources of ROS for vascular aging and their contribution to the increased cardiovascular risk in the elderly population with special emphasis on mitochondrial ROS formation and oxidative damage of mitochondrial DNA. Also the interaction (crosstalk) of mitochondria with nicotinamide adenosine dinucleotide phosphate (NADPH) oxidases is highlighted. Current concepts of vascular aging, consequences for the development of cardiovascular events and the particular role of ROS are evaluated on the basis of cell culture experiments, animal studies and clinical trials. Present data point to a more important role of oxidative stress for the maximal healthspan (healthy aging) than for the maximal lifespan.
Collapse
|
27
|
Tower J. Mitochondrial maintenance failure in aging and role of sexual dimorphism. Arch Biochem Biophys 2015; 576:17-31. [PMID: 25447815 PMCID: PMC4409928 DOI: 10.1016/j.abb.2014.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/08/2014] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Abstract
Gene expression changes during aging are partly conserved across species, and suggest that oxidative stress, inflammation and proteotoxicity result from mitochondrial malfunction and abnormal mitochondrial-nuclear signaling. Mitochondrial maintenance failure may result from trade-offs between mitochondrial turnover versus growth and reproduction, sexual antagonistic pleiotropy and genetic conflicts resulting from uni-parental mitochondrial transmission, as well as mitochondrial and nuclear mutations and loss of epigenetic regulation. Aging phenotypes and interventions are often sex-specific, indicating that both male and female sexual differentiation promote mitochondrial failure and aging. Studies in mammals and invertebrates implicate autophagy, apoptosis, AKT, PARP, p53 and FOXO in mediating sex-specific differences in stress resistance and aging. The data support a model where the genes Sxl in Drosophila, sdc-2 in Caenorhabditis elegans, and Xist in mammals regulate mitochondrial maintenance across generations and in aging. Several interventions that increase life span cause a mitochondrial unfolded protein response (UPRmt), and UPRmt is also observed during normal aging, indicating hormesis. The UPRmt may increase life span by stimulating mitochondrial turnover through autophagy, and/or by inhibiting the production of hormones and toxic metabolites. The data suggest that metazoan life span interventions may act through a common hormesis mechanism involving liver UPRmt, mitochondrial maintenance and sexual differentiation.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2910, United States.
| |
Collapse
|
28
|
Gonzalez-Freire M, de Cabo R, Bernier M, Sollott SJ, Fabbri E, Navas P, Ferrucci L. Reconsidering the Role of Mitochondria in Aging. J Gerontol A Biol Sci Med Sci 2015; 70:1334-42. [PMID: 25995290 DOI: 10.1093/gerona/glv070] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/23/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction has long been considered a major contributor to aging and age-related diseases. Harman's Mitochondrial Free Radical Theory of Aging postulated that somatic mitochondrial DNA mutations that accumulate over the life span cause excessive production of reactive oxygen species that damage macromolecules and impair cell and tissue function. Indeed, studies have shown that maximal oxidative capacity declines with age while reactive oxygen species production increases. Harman's hypothesis has been seriously challenged by recent studies showing that reactive oxygen species evoke metabolic health and longevity, perhaps through hormetic mechanisms that include autophagy. The purpose of this review is to scan the ever-growing literature on mitochondria from the perspective of aging research and try to identify priority questions that should be addressed in future research. METHODS A systematic search of peer-reviewed studies was performed using PubMed. Search terms included (i) mitochondria or mitochondrial; (ii) aging, ageing, older adults or elderly; and (iii) reactive oxygen species, mitochondria dynamics, mitochondrial proteostasis, cytosol, mitochondrial-associated membranes, redox homeostasis, electron transport chain, electron transport chain efficiency, epigenetic regulation, DNA heteroplasmy. RESULTS The importance of mitochondrial biology as a trait d'union between the basic biology of aging and the pathogenesis of age-related diseases is stronger than ever, although the emphasis has moved from reactive oxygen species production to other aspects of mitochondrial physiology, including mitochondrial biogenesis and turnover, energy sensing, apoptosis, senescence, and calcium dynamics. CONCLUSIONS Mitochondria could play a key role in the pathophysiology of aging or in the earlier stages of some events that lead to the aging phenotype. Therefore, mitochondria will increasingly be targeted to prevent and treat chronic diseases and to promote healthy aging.
Collapse
Affiliation(s)
| | | | | | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224
| | - Elisa Fabbri
- Translational Gerontology Branch, and Department of Medical and Surgical Sciences, University of Bologna, Italy 40126
| | - Placido Navas
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-CSIC, CIBERER, Instituto de Salud Carlos III, Sevilla, Spain 41013
| | | |
Collapse
|
29
|
Prigione A, Ruiz-Pérez MV, Bukowiecki R, Adjaye J. Metabolic restructuring and cell fate conversion. Cell Mol Life Sci 2015; 72:1759-77. [PMID: 25586562 PMCID: PMC11113500 DOI: 10.1007/s00018-015-1834-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/06/2015] [Accepted: 01/08/2015] [Indexed: 02/07/2023]
Abstract
Accumulating evidence implicates mitochondrial and metabolic pathways in the establishment of pluripotency, as well as in the control of proliferation and differentiation programs. From classic studies in mouse embryos to the latest findings in adult stem cells, human embryonic and induced pluripotent stem cells, an increasing number of evidence suggests that mitochondrial and metabolic-related processes might intertwine with signaling networks and epigenetic rewiring, thereby modulating cell fate decisions. This review summarizes the progresses in this exciting field of research. Dissecting these complex mitochondrial and metabolic mechanisms may lead to a more comprehensive understanding of stemness biology and to potential improvements in stem cell applications for biomedicine, cell therapy, and disease modeling.
Collapse
Affiliation(s)
- Alessandro Prigione
- Max Delbrueck Center for Molecular Medicine (MDC), Robert-Roessle-Str. 10, 13125, Berlin, Germany,
| | | | | | | |
Collapse
|
30
|
Müller M, Lu K, Reichert AS. Mitophagy and mitochondrial dynamics in Saccharomyces cerevisiae. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2766-74. [PMID: 25753536 DOI: 10.1016/j.bbamcr.2015.02.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/10/2015] [Accepted: 02/27/2015] [Indexed: 12/13/2022]
Abstract
Mitochondria fulfill central cellular functions including energy metabolism, iron-sulfur biogenesis, and regulation of apoptosis and calcium homeostasis. Accumulation of dysfunctional mitochondria is observed in ageing and many human diseases such as cancer and various neurodegenerative disorders. Appropriate quality control of mitochondria is important for cell survival in most eukaryotic cells. One important pathway in this respect is mitophagy, a selective form of autophagy which removes excess and dysfunctional mitochondria. In the past decades a series of essential factors for mitophagy have been identified and characterized. However, little is known about the molecular mechanisms regulating mitophagy. The role of mitochondrial dynamics in mitophagy is controversially discussed. Here we will review recent advances in this context promoting our understanding on the molecular regulation of mitophagy in Saccharomyces cerevisiae and on the role of mitochondrial dynamics in mitochondrial quality control.
Collapse
Affiliation(s)
- Matthias Müller
- Mitochondrial Biology, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Germany; Mitochondrial Biology, Medical School, Goethe University Frankfurt am Main, Germany
| | - Kaihui Lu
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Andreas S Reichert
- Mitochondrial Biology, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Germany; Mitochondrial Biology, Medical School, Goethe University Frankfurt am Main, Germany; Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
31
|
Shadyab AH, LaCroix AZ. Genetic factors associated with longevity: a review of recent findings. Ageing Res Rev 2015; 19:1-7. [PMID: 25446805 DOI: 10.1016/j.arr.2014.10.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/20/2014] [Accepted: 10/27/2014] [Indexed: 12/21/2022]
Abstract
Given the rising rate of survival into advanced old age in the United States, achieving longevity and healthy aging is becoming increasingly important. Besides maintaining healthy lifestyle behaviors, positive aging outcomes may also be heritable, with estimates ranging from 20% to 35%. In this qualitative review, we summarize recent findings on genetic factors linked to longevity across different populations and study designs. Recent studies not only confirm the association of APOE with longevity in different populations, but also implicate several other pathways that may influence longevity including nitric oxide production, inflammation, immunity, and DNA damage response and repair. Recent evidence also suggests that mitochondrial DNA may play an important role in attaining longevity. Despite these implicated pathways, longevity may be a polygenic trait influenced by a complex interplay of multiple genes. Future genetic studies on aging would benefit from larger samples of long-lived individuals, birth-cohort matched controls, inclusion of different aging phenotypes (e.g., aging free of morbidities), and analysis of gender differences.
Collapse
Affiliation(s)
- Aladdin H Shadyab
- San Diego State University/University of California, San Diego Joint-Doctoral Program in Public Health (Epidemiology), USA; Graduate School of Public Health, San Diego State University, Hardy Tower Room 119, 5500 Campanile Drive, San Diego, CA, 92182-4162, USA.
| | - Andrea Z LaCroix
- Department of Family and Preventive Medicine, University of California, San Diego School of Medicine, La Jolla, CA, USA.
| |
Collapse
|