1
|
Monesi N, Fernandes GM, Valer FB, Uliana JVC, Trinca V, Azzolini AECS, Gorab E, Alberici LC. Identification and characterization of a laterally transferred alternative oxidase (AOX) in a terrestrial insect, the dipteran Pseudolycoriella hygida. Biochimie 2025; 233:60-74. [PMID: 39988053 DOI: 10.1016/j.biochi.2025.02.007] [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: 07/07/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Alternative oxidase (AOX) (EC 1.10.3.11) is a terminal oxidase in the mitochondrial inner membrane that branches the canonical electron transport system (ETS). AOX is ubiquitous in plants, frequently found in fungi and protists and presents a more sporadic distribution in metazoans. More recently, AOX has gained attention due to its potential application in gene therapy for treatment of mitochondrial diseases. Here we characterized the AOX in the basal Dipteran, Pseudolycoriella hygida using a combination of genomic analyses, molecular, functional and in vivo survival assays. AOX is a single copy gene that encodes three developmental stage specific protein isoforms. AOX localizes to the mitochondria in adult thoracic muscles, which present cyanide-resistant respiration that is sensitive to the AOX inhibitor salicylhydroxamic acid (SHAM). Both the cyanide-resistant respiration and AOX levels gradually increase during aging, but are not influenced by thermal stress. Thoracic mitochondria respire using substrates derived from several metabolic routes, such as pyruvate, proline, acylcarnitine, NADH and glycerol-3P, and present values of oxidative phosphorylation capacity ((P-L)/E = 0.70) and coupling (P/L = 4.35; L/E = 0.21). Adult flies exhibit a high survival resistance for SHAM-sensitive complex III inhibition. Together, our results demonstrate the presence of a functional AOX in a terrestrial arthropod and provide insights regarding AOX function in animals and evolution of respiratory systems in metazoans. Psl. hygida emerges as a natural and valuable model for comprehensive AOX research at the whole-organism level which complements models expressing the heterologous enzyme.
Collapse
Affiliation(s)
- Nadia Monesi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida Professor Doutor Zeferino Vaz, s/n. Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| | - Guilherme Magre Fernandes
- Programa de Biologia Celular e Molecular, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo. Av. Bandeirantes, 3900, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil.
| | - Felipe Berti Valer
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida Professor Doutor Zeferino Vaz, s/n. Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| | - João Vítor Cardoso Uliana
- Programa de Biologia Celular e Molecular, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo. Av. Bandeirantes, 3900, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil.
| | - Vitor Trinca
- Programa de Biologia Celular e Molecular, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo. Av. Bandeirantes, 3900, Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil.
| | - Ana Elisa Caleiro Seixas Azzolini
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida Professor Doutor Zeferino Vaz, s/n. Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| | - Eduardo Gorab
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo. Rua do Matão, trav. 14, nº 321, Cidade Universitária, São Paulo, SP, 05508-090, Brazil.
| | - Luciane Carla Alberici
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Avenida Professor Doutor Zeferino Vaz, s/n. Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
2
|
Luo L, Wang M, Liu Y, Li J, Bu F, Yuan H, Tang R, Liu C, He G. Sequencing and characterizing human mitochondrial genomes in the biobank-based genomic research paradigm. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1610-1625. [PMID: 39843848 DOI: 10.1007/s11427-024-2736-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/18/2024] [Indexed: 01/24/2025]
Abstract
Human mitochondrial DNA (mtDNA) harbors essential mutations linked to aging, neurodegenerative diseases, and complex muscle disorders. Due to its uniparental and haploid inheritance, mtDNA captures matrilineal evolutionary trajectories, playing a crucial role in population and medical genetics. However, critical questions about the genomic diversity patterns, inheritance models, and evolutionary and medical functions of mtDNA remain unresolved or underexplored, particularly in the transition from traditional genotyping to large-scale genomic analyses. This review summarizes recent advancements in data-driven genomic research and technological innovations that address these questions and clarify the biological impact of nuclear-mitochondrial segments (NUMTs) and mtDNA variants on human health, disease, and evolution. We propose a streamlined pipeline to comprehensively identify mtDNA and NUMT genomic diversity using advanced sequencing and computational technologies. Haplotype-resolved mtDNA sequencing and assembly can distinguish authentic mtDNA variants from NUMTs, reduce diagnostic inaccuracies, and provide clearer insights into heteroplasmy patterns and the authenticity of paternal inheritance. This review emphasizes the need for integrative multi-omics approaches and emerging long-read sequencing technologies to gain new insights into mutation mechanisms, the influence of heteroplasmy and paternal inheritance on mtDNA diversity and disease susceptibility, and the detailed functions of NUMTs.
Collapse
Affiliation(s)
- Lintao Luo
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China
| | - Mengge Wang
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China.
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China.
- Anti-Drug Technology Center of Guangdong Province, Guangzhou, 510230, China.
| | - Yunhui Liu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China
| | - Jianbo Li
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China
| | - Fengxiao Bu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China
| | - Huijun Yuan
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China.
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China.
| | - Renkuan Tang
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China.
| | - Chao Liu
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China.
- Anti-Drug Technology Center of Guangdong Province, Guangzhou, 510230, China.
| | - Guanglin He
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China.
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China.
- Anti-Drug Technology Center of Guangdong Province, Guangzhou, 510230, China.
| |
Collapse
|
3
|
Kathiresan DS, Balasubramani R, Marudhachalam K, Jaiswal P, Ramesh N, Sureshbabu SG, Puthamohan VM, Vijayan M. Role of Mitochondrial Dysfunctions in Neurodegenerative Disorders: Advances in Mitochondrial Biology. Mol Neurobiol 2025; 62:6827-6855. [PMID: 39269547 DOI: 10.1007/s12035-024-04469-x] [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: 04/04/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
Mitochondria, essential organelles responsible for cellular energy production, emerge as a key factor in the pathogenesis of neurodegenerative disorders. This review explores advancements in mitochondrial biology studies that highlight the pivotal connection between mitochondrial dysfunctions and neurological conditions such as Alzheimer's, Parkinson's, Huntington's, ischemic stroke, and vascular dementia. Mitochondrial DNA mutations, impaired dynamics, and disruptions in the ETC contribute to compromised energy production and heightened oxidative stress. These factors, in turn, lead to neuronal damage and cell death. Recent research has unveiled potential therapeutic strategies targeting mitochondrial dysfunction, including mitochondria targeted therapies and antioxidants. Furthermore, the identification of reliable biomarkers for assessing mitochondrial dysfunction opens new avenues for early diagnosis and monitoring of disease progression. By delving into these advancements, this review underscores the significance of understanding mitochondrial biology in unraveling the mechanisms underlying neurodegenerative disorders. It lays the groundwork for developing targeted treatments to combat these devastating neurological conditions.
Collapse
Affiliation(s)
- Divya Sri Kathiresan
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Rubadevi Balasubramani
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Kamalesh Marudhachalam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Piyush Jaiswal
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Nivedha Ramesh
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Suruthi Gunna Sureshbabu
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India
| | - Vinayaga Moorthi Puthamohan
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Nadu, Tamil, 641046, India.
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
4
|
Moura JP, Oliveira PJ, Urbano AM. Mitochondria: An overview of their origin, genome, architecture, and dynamics. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167803. [PMID: 40118291 DOI: 10.1016/j.bbadis.2025.167803] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Mitochondria are traditionally viewed as the powerhouses of eukaryotic cells, i.e., the main providers of the metabolic energy required to maintain their viability and function. However, the role of these ubiquitous intracellular organelles far extends energy generation, encompassing a large suite of functions, which they can adjust to changing physiological conditions. These functions rely on a sophisticated membrane system and complex molecular machineries, most of which imported from the cytosol through intricate transport systems. In turn, mitochondrial plasticity is rooted on mitochondrial biogenesis, mitophagy, fusion, fission, and movement. Dealing with all these aspects and terminology can be daunting for newcomers to the field of mitochondria, even for those with a background in biological sciences. The aim of the present educational article, which is part of a special issue entitled "Mitochondria in aging, cancer and cell death", is to present these organelles in a simple and concise way. Complex molecular mechanisms are deliberately omitted, as their inclusion would defeat the stated purpose of the article. Also, considering the wide scope of the article, coverage of each topic is necessarily limited, with the reader directed to excellent reviews, in which the different topics are discussed in greater depth than is possible here. In addition, the multiple cell type-specific genotypic and phenotypic differences between mitochondria are largely ignored, focusing instead on the characteristics shared by most of them, with an emphasis on mitochondria from higher eukaryotes. Also ignored are highly degenerate mitochondrion-related organelles, found in various anaerobic microbial eukaryotes lacking canonical mitochondria.
Collapse
Affiliation(s)
- João P Moura
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| | - Ana M Urbano
- Molecular Physical-Chemistry R&D Unit, Centre for Investigation in Environment, Genetics and Oncobiology (CIMAGO), Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
5
|
Yu Y, Zhang L, Wen S, Li C, Tan L, Wu X, Zou Y, Liu T. Evaluation of visual function and morphological changes in patients with adrenoleukodystrophy using SS-OCT and PVEP. Doc Ophthalmol 2025:10.1007/s10633-025-10021-6. [PMID: 40347222 DOI: 10.1007/s10633-025-10021-6] [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: 12/02/2024] [Accepted: 04/11/2025] [Indexed: 05/12/2025]
Abstract
PURPOSE This study aims to evaluate visual function and morphological changes in patients with adrenoleukodystrophy (ALD) through sweep-source optical coherence tomography (SS-OCT) and patterned visual evoked potentials (PVEP), with the goal of providing more accurate and comprehensive data for the diagnosis, monitoring, and assessment of treatment efficacy in patients with ALD. METHODS This study included 46 ALD patients and 44 healthy controls. The thickness of the retinal nerve fiber layer (RNFL) around the optic disc, as well as the macular RNFL and ganglion cell-inner plexiform layer (GCIPL) thickness, was assessed using SS-OCT. The peak time and amplitude of the P100 wave were also measured using PVEP. Factors that were statistically significant in the univariate analysis were subjected to stepwise binary logistic regression analysis for further investigation. Predictive performance was evaluated by constructing receiver operating characteristic (ROC) curves and compared using DeLong's test. RESULTS Compared with the control patients, ALD patients presented a significant increase in the peak time of the P100 wave (P < 0.05); however, no notable difference in amplitude was observed (P > 0.05). Additionally, substantial decreases in the RNFL and GCIPL thicknesses were observed within the parafoveal ring, especially in the superior and nasal quadrants (P < 0.05). The area under the curve for the binary logistic stepwise regression model was 0.883, with a sensitivity of 0.95, which surpassed the performance of the individual parameters. CONCLUSION ALD patients present with abnormal retinal structures and a PVEP peak time delay. Combining these two parameters could increase the accuracy of an early ALD diagnosis.
Collapse
Affiliation(s)
- Yongqiu Yu
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Lujie Zhang
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Shuiqian Wen
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Chongyi Li
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xian Wu
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Yangcheng Zou
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Ting Liu
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
| |
Collapse
|
6
|
Semo D, Shomanova Z, Sindermann J, Mohr M, Evers G, Motloch LJ, Reinecke H, Godfrey R, Pistulli R. Persistent Monocytic Bioenergetic Impairment and Mitochondrial DNA Damage in PASC Patients with Cardiovascular Complications. Int J Mol Sci 2025; 26:4562. [PMID: 40429707 PMCID: PMC12111130 DOI: 10.3390/ijms26104562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiovascular complications are a hallmark of Post-Acute Sequelae of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection (PASC), yet the mechanisms driving persistent cardiac dysfunction remain poorly understood. Emerging evidence implicates mitochondrial dysfunction in immune cells as a key contributor. This study investigated whether CD14++ monocytes from long COVID patients exhibit bioenergetic impairment, mitochondrial DNA (mtDNA) damage, and defective oxidative stress adaptation, which may underlie cardiovascular symptoms in PASC. CD14++ monocytes were isolated from 14 long COVID patients with cardiovascular symptoms (e.g., dyspnea, angina) and 10 age-matched controls with similar cardiovascular risk profiles. Mitochondrial function was assessed using a Seahorse Agilent Analyzer under basal conditions and after oxidative stress induction with buthionine sulfoximine (BSO). Mitochondrial membrane potential was measured via Tetramethylrhodamine Ethyl Ester (TMRE) assay, mtDNA integrity via qPCR, and reactive oxygen species (ROS) dynamics via Fluorescence-Activated Cell Sorting (FACS). Parallel experiments exposed healthy monocytes to SARS-CoV-2 spike protein to evaluate direct viral effects. CD14++ monocytes from long COVID patients with cardiovascular symptoms (n = 14) exhibited profound mitochondrial dysfunction compared to age-matched controls (n = 10). Under oxidative stress induced by buthionine sulfoximine (BSO), long COVID monocytes failed to upregulate basal respiration (9.5 vs. 30.4 pmol/min in controls, p = 0.0043), showed a 65% reduction in maximal respiration (p = 0.4035, ns) and demonstrated a 70% loss of spare respiratory capacity (p = 0.4143, ns) with significantly impaired adaptation to BSO challenge (long COVID + BSO: 9.9 vs. control + BSO: 54 pmol/min, p = 0.0091). Proton leak, a protective mechanism against ROS overproduction, was blunted in long COVID monocytes (3-fold vs. 13-fold elevation in controls, p = 0.0294). Paradoxically, long COVID monocytes showed reduced ROS accumulation after BSO treatment (6% decrease vs. 1.2-fold increase in controls, p = 0.0015) and elevated mitochondrial membrane potential (157 vs. 113.7 TMRE fluorescence, p = 0.0179), which remained stable under oxidative stress. mtDNA analysis revealed severe depletion (80% reduction, p < 0.001) and region-specific damage, with 75% and 70% reductions in amplification efficiency for regions C and D (p < 0.05), respectively. In contrast, exposure of healthy monocytes to SARS-CoV-2 spike protein did not recapitulate these defects, with preserved basal respiration, ATP production, and spare respiratory capacity, though coupling efficiency under oxidative stress was reduced (p < 0.05). These findings suggest that mitochondrial dysfunction in long COVID syndrome arises from maladaptive host responses rather than direct viral toxicity, characterized by bioenergetic failure, impaired stress adaptation, and mitochondrial genomic instability. This study identifies persistent mitochondrial dysfunction in long COVID monocytes as a critical driver of cardiovascular complications in PASC. Key defects-bioenergetic failure, impaired stress adaptation and mtDNA damage-correlate with clinical symptoms like heart failure and exercise intolerance. The stable elevation of mitochondrial membrane potential and resistance to ROS induction suggest maladaptive remodeling of mitochondrial physiology. These findings position mitochondrial resilience as a therapeutic target, with potential strategies including antioxidants, mtDNA repair agents or metabolic modulators. The dissociation between spike protein exposure and mitochondrial dysfunction highlights the need to explore host-directed mechanisms in PASC pathophysiology. This work advances our understanding of long COVID cardiovascular sequelae and provides a foundation for biomarker development and targeted interventions to mitigate long-term morbidity.
Collapse
Affiliation(s)
- Dilvin Semo
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany;
- Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany;
| | - Zornitsa Shomanova
- Interdisciplinary Heart Failure Section, Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany; (Z.S.); (J.S.)
| | - Jürgen Sindermann
- Interdisciplinary Heart Failure Section, Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany; (Z.S.); (J.S.)
| | - Michael Mohr
- Department of Medicine A, Hematology, Oncology, and Pulmonary Medicine, University Hospital Münster, 48149 Münster, Germany; (M.M.); (G.E.)
| | - Georg Evers
- Department of Medicine A, Hematology, Oncology, and Pulmonary Medicine, University Hospital Münster, 48149 Münster, Germany; (M.M.); (G.E.)
| | - Lukas J. Motloch
- Department of Internal Medicine II, Paracelsus Medical University, 5020 Salzburg, Austria;
- Department of Internal Medicine II, Salzkammergut Klinikum, OÖG, 4840 Vöcklabruck, Austria
- Department of Cardiology, Kepler University Hospital, Medical Faculty, Johannes Kepler University, 4020 Linz, Austria
| | - Holger Reinecke
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany;
- Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany;
- Interdisciplinary Heart Failure Section, Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany; (Z.S.); (J.S.)
| | - Rinesh Godfrey
- Vascular Signalling, Molecular Cardiology, Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany;
| | - Rudin Pistulli
- Department of Cardiology I, Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, 48149 Münster, Germany;
| |
Collapse
|
7
|
Tamashiro H, Ishikawa K, Sadotomo K, Ogasawara E, Nakada K. Mitochondrial Respiratory Dysfunction Is Not Correlated With Mitochondrial Genotype in Premature Aging Mice. Aging Cell 2025:e70085. [PMID: 40318128 DOI: 10.1111/acel.70085] [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: 12/28/2024] [Revised: 03/17/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025] Open
Abstract
mtDNA mutator mice (Polgmut/mut mice) have reinforced the mitochondrial theory of aging. These mice accumulate multiple mutations in mtDNA with age due to a homozygous proofreading-deficient mutation in mtDNA polymerase gamma (Polg), resulting in mitochondrial respiratory dysfunction and premature aging phenotypes. However, whether the accumulation of multiple mutations in Polgmut/mut mice induces mitochondrial respiratory dysfunction remains unclear. Here, we determined the accurate mtDNA genotype, including the frequency of total mutations and the number of non-synonymous substitutions and pathogenic mutations, using next-generation sequencing in the progeny of all three genotypes obtained from the mating of heterozygous mtDNA mutator mice (Polg+/mut mice) and examined their correlation with mitochondrial respiratory activity. Although Polg+/mut mice showed equivalent mtDNA genotype to Polg+/+ (wild-type) mice, the mitochondrial respiratory activity in the Polg+/mut mice was mildly reduced. To further investigate the causal relationship between mtDNA genotype and mitochondrial respiratory activity, we experimentally varied the mtDNA genotype in Polg mice. However, mitochondrial respiratory activity was mildly reduced in Polg+/mut mice and severely reduced in Polgmut/mut mice, regardless of the mtDNA genotype. Moreover, by varying the mtDNA genotype, some Polg+/+ mice showed mtDNA genotype equivalent to those of Polgmut/mut mice, but mitochondrial respiratory activity in Polg+/+ mice was normal. These results indicate that the mitochondrial respiratory dysfunction observed in mice with proofreading-deficient mutation in Polg is correlated with the nuclear genotype of Polg rather than the mtDNA genotype. Thus, the mitochondrial theory of aging in Polgmut/mut mice needs further re-examination.
Collapse
Affiliation(s)
- Hiroaki Tamashiro
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, Japan
| | - Kaori Ishikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| | - Koichi Sadotomo
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, Japan
| | - Emi Ogasawara
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Kazuto Nakada
- Institute of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
8
|
Alves JL, Quinta-Ferreira RM, Quinta-Ferreira ME, Matias CM. Exploring different mechanisms of reactive oxygen species formation in hypoxic conditions at the hippocampal CA3 area. Mol Cell Endocrinol 2025; 601:112517. [PMID: 40054836 DOI: 10.1016/j.mce.2025.112517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/17/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Hypoxia can lead to severe consequences for brain function, particularly in regions with high metabolic demands such as the hippocampus. Excessive production of reactive oxygen species (ROS) during hypoxia can initiate a cascade of oxidative stress, evoking cellular damage and neuronal dysfunction. Most of the studies characterizing the formation of ROS are performed in the context of ischemia induced by oxygen-glucose deprivation, thus, the role of hypoxia in less severe conditions requires further clarification. The aim of this work was to identify the major mechanisms of ROS generation and assess flavoprotein autofluorescence changes. For ROS detection, the slices were incubated with the indicator H2DCFDA, while intrinsic FAD-linked autofluorescence was recorded from indicator free slices. All signals were measured under hypoxia, at the hippocampal mossy fiber synapses of CA3 area, which were chemically stimulated using 20 mM KCl. The results suggest that ROS is formed in the mitochondria, during moderate hypoxia. The blockage of mitochondrial complexes I, III and IV with rotenone, myxothiazol and sodium azide, respectively, and of the mitochondrial calcium uniporter with Ru265, led to the abolishment of ROS changes and to an increase of FAD-linked autofluorescence (with the exception of the complexes III and IV). The blockage of the enzyme oxidases NADPH and xanthine oxidase also impaired ROS formation and rose FAD-linked autofluorescence. Thus, the blockage of any of the steps of the process of ROS formation, namely the activation of critical MRC complexes, calcium entry into the mitochondria, or enzyme oxidases activity, ceases the production of ROS.
Collapse
Affiliation(s)
- João L Alves
- Department of Life Sciences, University of Coimbra, Portugal; CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Rosa M Quinta-Ferreira
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - M Emília Quinta-Ferreira
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, University of Coimbra, Portugal
| | - Carlos M Matias
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, UTAD, Vila Real, Portugal.
| |
Collapse
|
9
|
Kui L, Ma P, Zhao W, Yan B, Kuang X, Li B, Geng R, Zheng T, Zheng Q. Developmental cochlear defects are involved in early-onset hearing loss in A/J mice. Dev Dyn 2025; 254:436-449. [PMID: 39291400 DOI: 10.1002/dvdy.741] [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: 04/27/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND A/J mice exhibited a severe hearing loss (HL) at juvenile stage. Up-to-date, studies on HL in A/J mice have mostly focused on the damage or dysfunction of hair cells (HCs), spiral ganglion neurons (SGNs), and stereocilia. We examined A/J mice at the early postnatal stage and found that the damage and the loss of outer hair cells (OHCs) are not severe enough to explain the profound HL observed at this age, which suggests that other cochlear defects may be responsible for HL. To better understand the mechanisms of early-onset HLin A/J mice, we characterized the pathology of the cochlea from postnatal day 3 to day 21. RESULTS Our results showed defects in cochlear HC stereocilia and MET channel function as early as 3 days old. We also found abnormal localization and a significant reduction in the number of ribbon synapses in 2-week-old A/J mice. There are also abnormalities in the cochlear nerve innervation and terminal swellings in 3-week-old A/J mice. CONCLUSION All of the abnormalities of cochlear existed in the A/J mice were identified in the juvenile stage and occurred before HCs or auditory nerve loss and was the initial pathological change. Our results suggest that developmental defects and subsequent cochlear degeneration are responsible for early-onset hearing loss in A/J mice.
Collapse
Affiliation(s)
- Lihong Kui
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Peng Ma
- Department of Medical Genetics and Cell Biology, Binzhou Medical University, Yantai, China
| | - Wenben Zhao
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Bin Yan
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Xiaojing Kuang
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Bo Li
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Ruishuang Geng
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Tihua Zheng
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| | - Qingyin Zheng
- Hearing and Speech Rehabilitation Institute, College of Special Education and Rehabilitation, Binzhou Medical University, Yantai, China
| |
Collapse
|
10
|
Antico O, Thompson PW, Hertz NT, Muqit MMK, Parton LE. Targeting mitophagy in neurodegenerative diseases. Nat Rev Drug Discov 2025; 24:276-299. [PMID: 39809929 DOI: 10.1038/s41573-024-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Mitochondrial dysfunction is a hallmark of idiopathic neurodegenerative diseases, including Parkinson disease, amyotrophic lateral sclerosis, Alzheimer disease and Huntington disease. Familial forms of Parkinson disease and amyotrophic lateral sclerosis are often characterized by mutations in genes associated with mitophagy deficits. Therefore, enhancing the mitophagy pathway may represent a novel therapeutic approach to targeting an underlying pathogenic cause of neurodegenerative diseases, with the potential to deliver neuroprotection and disease modification, which is an important unmet need. Accumulating genetic, molecular and preclinical model-based evidence now supports targeting mitophagy in neurodegenerative diseases. Despite clinical development challenges, small-molecule-based approaches for selective mitophagy enhancement - namely, USP30 inhibitors and PINK1 activators - are entering phase I clinical trials for the first time.
Collapse
Affiliation(s)
- Odetta Antico
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul W Thompson
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK
| | | | - Miratul M K Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura E Parton
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
11
|
Heiduschka S, Prigione A. iPSC models of mitochondrial diseases. Neurobiol Dis 2025; 207:106822. [PMID: 39892770 DOI: 10.1016/j.nbd.2025.106822] [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: 06/04/2024] [Revised: 11/17/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025] Open
Abstract
Mitochondrial diseases are historically difficult to study. They cause multi-systemic defects with prevalent impairment of hard-to-access tissues such as the brain and the heart. Furthermore, they suffer from a paucity of conventional model systems, especially because of the challenges associated with mitochondrial DNA (mtDNA) engineering. Consequently, most mitochondrial diseases are currently untreatable. Human induced pluripotent stem cells (iPSCs) represent a promising approach for developing human model systems and assessing therapeutic avenues in a patient- and tissue-specific context. iPSCs are being increasingly used to investigate mitochondrial diseases, either for dissecting mutation-specific defects within two-dimensional (2D) or three-dimensional (3D) progenies or for unveiling the impact of potential treatment options. Here, we review how iPSC-derived 2D cells and 3D organoid models have been applied to the study of mitochondrial diseases caused by either nuclear or mtDNA defects. We anticipate that the field of iPSC-driven modeling of mitochondrial diseases will continue to grow, likely leading to the development of innovative platforms for treatment discovery and toxicity that could benefit the patient community suffering from these debilitating disorders with highly unmet medical needs.
Collapse
Affiliation(s)
- Sonja Heiduschka
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Germany; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany.
| |
Collapse
|
12
|
Peng W, Tai W, Li B, Wang H, Wang T, Guo S, Zhang X, Dong P, Tian C, Feng S, Yang L, Cheng G, Zheng B. Inhalable nanocatalytic therapeutics for viral pneumonia. NATURE MATERIALS 2025; 24:637-648. [PMID: 39592721 DOI: 10.1038/s41563-024-02041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 10/04/2024] [Indexed: 11/28/2024]
Abstract
Pneumonia is a ubiquitous disease caused by viral and bacterial infections, characterized by high levels of reactive oxygen species in inflamed areas. Therapeutic strategies targeting reactive oxygen species levels in pneumonia have limited success due to the intricate nature of lung tissues and lung inflammatory responses. Here we describe an inhalable, non-invasive therapeutic platform composed of engineered cerium-based tannic acid nanozymes bound to a self-assembling peptide. In vitro and in vivo studies show that the nanozyme is internalized mostly by activated macrophages and epithelial cells in the inflamed sites. In the oxidative environments of a mouse model of viral pneumonia, nanozyme aggregates into catalytically active structures that reduce reactive oxygen species levels and inflammatory cytokine production and promote macrophage polarization to the prohealing (M2) phenotype. Moreover, the nanozyme attenuates bacterial inflammation and reduces tissue damage in a mouse viral pneumonia model with secondary bacterial infection. Overall, this nanozyme platform is a promising strategy for treating pneumonia and its associated conditions.
Collapse
Affiliation(s)
- Wenchang Peng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Wanbo Tai
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Bowen Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Hua Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Tao Wang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Shuyue Guo
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Xu Zhang
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Pengyuan Dong
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Chongyu Tian
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Shengyong Feng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Long Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China.
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, China.
- Southwest United Graduate School, Kunming, China.
| | - Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
13
|
Cao ML, Han RY, Chen SD, Zhao DY, Shi MY, Zou JH, Li L, Jiang HK. Gene Editing: An Effective Tool for the Future Treatment of Kidney Disease. J Inflamm Res 2025; 18:4001-4018. [PMID: 40125088 PMCID: PMC11927957 DOI: 10.2147/jir.s506760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Gene editing technology involves modifying target genes to alter genetic traits and generate new phenotypes. Beginning with zinc-finger nucleases (ZFN) and transcription activator-like effector nucleases (TALEN), the field has evolved through the advent of clustered regularly interspaced short palindromic repeats and CRISPR-associated protein (CRISPR-Cas) systems, and more recently to base editors (BE) and prime editors (PE). These innovations have provided deep insights into the molecular mechanisms of complex biological processes and have paved the way for novel therapeutic strategies for a range of diseases. Gene editing is now being applied in the treatment of both genetic and acquired kidney diseases, as well as in kidney transplantation and the correction of genetic mutations. This review explores the current applications of mainstream gene editing technologies in biology, with a particular emphasis on their roles in kidney disease research and treatment of. It also addresses the limitations and challenges associated with these technologies, while offering perspectives on their future potential in this field.
Collapse
Affiliation(s)
- Mei-Ling Cao
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Rui-Yi Han
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Si-Da Chen
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People’s Republic of China
| | - Dan-Yang Zhao
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Ming-Yue Shi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Jia-Hui Zou
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| | - Lei Li
- Department of Orthopaedic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People’s Republic of China
| | - Hong-Kun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, People’s Republic of China
| |
Collapse
|
14
|
Hanada S, Ishikawa K, Shirai T, Takemasa T, Nakada K. Endurance swimming exacerbates mitochondrial myopathy in mice with high mtDNA deletions. Mitochondrion 2025; 81:102010. [PMID: 39956167 DOI: 10.1016/j.mito.2025.102010] [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: 12/09/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Recent studies have reported that endurance exercise enhances mitochondrial function, facilitating discussions of its potential as a therapeutic strategy for mitochondrial diseases caused by the accumulation of mutant mitochondrial DNA (mtDNA). In this study, we assessed the effects of endurance exercise on muscle pathology in a mitochondrial disease mouse model (mito-miceΔ) that is characterized by severe clinical phenotypes owing to the predominant accumulation of mtDNA with a large-scale deletion (ΔmtDNA). Contrary to expectations that endurance exercise may enhance mitochondrial function, endurance exercise exacerbated muscle pathology in mito-miceΔ. Therefore, exercise interventions should be potentially avoided in patients with severe mitochondrial diseases.
Collapse
Affiliation(s)
- Sho Hanada
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan
| | - Kaori Ishikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.
| | - Takanaga Shirai
- Institute of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan; Research Fellow of Japan Society for Promotion Science, Chiyoda-ku, Tokyo 102-0083, Japan; Department of Human Sciences, Kanagawa University, 3-27-1 Rokkakubashi, Kanagawa-ku, Yokohama-shi, Kanagawa 221-8686, Japan
| | - Tohru Takemasa
- Institute of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
| | - Kazuto Nakada
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.
| |
Collapse
|
15
|
Zhang X, Zhang L, Xiang W. The impact of mitochondrial dysfunction on ovarian aging. J Transl Med 2025; 23:211. [PMID: 39980008 PMCID: PMC11844166 DOI: 10.1186/s12967-025-06223-w] [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: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
IMPORTANCE Ovarian aging has become a focal point in current research on female aging and refers to the gradual decline in ovarian function as women age. Numerous factors influence ovarian aging, among which mitochondrial function is one because it plays a crucial role by affecting oocytes and granulosa cells. Mitochondrial deterioration not only leads to a decrease in oocyte quality but also hinders follicle development, further impacting women's reproductive health and fertility. OBJECTIVE This review summarizes and integrates research on the impact of mitochondrial function on ovarian aging, outlining the mechanisms by which mitochondria regulate the functions of oocytes and granulosa cells. This study aims to provide potential therapeutic directions to mitigate mitochondrial decline and support female reproductive health. EVIDENCE REVIEW According to a 2023 study published in Cell, factors such as oxidative stress, mitochondrial dysfunction, chronic inflammation, and telomere shortening collectively drive ovarian aging, directly affecting female fertility. Among these factors, mitochondrial dysfunction plays a key role. This study reviewed literature from databases such as PubMed, Google Scholar, and CNKI, using keywords such as "mitochondrial dysfunction", "decline in oocyte quality and quantity", and "ovarian aging", aiming to summarize current research on the mechanisms of the impact of mitochondrial dysfunction on ovarian aging and provide theoretical support for future exploration of related therapeutic strategies. FINDINGS The main characteristics of ovarian aging include a decline in oocyte quantity and quality, fluctuations in hormone levels, and a reduction in granulosa cell function. Studies have shown that mitochondria affect fertility by regulating cellular energy metabolism, exacerbating oxidative stress, causing mitochondrial DNA (mtDNA) damage, and impacting the physiological function of granulosa cells within the ovary, gradually diminishing the ovarian reserve. CONCLUSION This review focuses on analyzing the effects of mitochondrial decline on energy production in oocytes and granulosa cells, the accumulation of reactive oxygen species (ROS), and the calcium ion (Ca2+) concentration, which all contribute to the ovarian aging process, and understanding them will provide new insights into the mechanisms of ovarian aging. RELEVANCE Therapeutic interventions targeting mitochondrial dysfunction may help delay ovarian aging and improve female reproductive health.
Collapse
Affiliation(s)
- Xiaoyue Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Wenpei Xiang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
16
|
Sato N, Kusano T, Nagata K, Okamoto K. A non-purine inhibitor of xanthine oxidoreductase mitigates adenosine triphosphate degradation under hypoxic conditions in mouse brain. Brain Res 2025; 1849:149444. [PMID: 39755194 DOI: 10.1016/j.brainres.2025.149444] [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: 07/22/2024] [Revised: 12/12/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
The brain is an organ that consumes a substantial amount of oxygen, and a reduction in oxygen concentration can rapidly lead to significant and irreversible brain injury. The progression of brain injury during hypoxia involves the depletion of intracellular adenosine triphosphate (ATP) due to decreased oxidative phosphorylation in the inner mitochondrial membrane. Allopurinol is a purine analog inhibitor of xanthine oxidoreductase that protects against hypoxic/ischemic brain injury; however, its underlying mechanism of action remains unclear. In addition, febuxostat is a non-purine xanthine oxidoreductase inhibitor with a different inhibitory mechanism from allopurinol. The impact of febuxostat on brain injury has not been well investigated. Therefore, this study aimed to examine brain ATP and its catabolite levels in the presence or absence of allopurinol and febuxostat under hypoxic conditions by inactivating brain metabolism using focal microwave irradiation. The hypoxic treatment caused a decrease in the adenylate energy charge and ATP levels and an increase in its catabolic products in mouse brains. The febuxostat group showed higher energy charge and ATP levels and lower ATP catabolites than the control group. Notably, despite the comparable suppression of uric acid production in both inhibitor groups, allopurinol treatment was less effective than febuxostat. These results suggest that febuxostat effectively prevents hypoxia-induced ATP degradation in the brain and that its effect is more potent than allopurinol. This study will contribute to developing therapies for improving hypoxia-induced brain dysfunction.
Collapse
Affiliation(s)
- Nana Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Teruo Kusano
- Department of Biochemistry and Molecular Biology, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-Ku, Tokyo, Japan
| | - Koji Nagata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Ken Okamoto
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
17
|
Yen K, Miller B, Kumagai H, Silverstein A, Cohen P. Mitochondrial-derived microproteins: from discovery to function. Trends Genet 2025; 41:132-145. [PMID: 39690001 PMCID: PMC11794013 DOI: 10.1016/j.tig.2024.11.010] [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: 07/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024]
Abstract
Given the uniqueness of the mitochondria, and the fact that they have their own genome, mitochondrial-derived microproteins (MDPs) are similar to, but different from, nuclear-encoded microproteins. The discovery of an increasing number of microproteins from this organelle and the importance of mitochondria to cellular and organismal health make it a priority to study this novel class of proteins in search of possible therapeutic targets and cures. In this review, we discuss the history of MDP discovery, describe the function of each MDP, and conclude with future goals and techniques to help discover more MDPs.
Collapse
Affiliation(s)
- Kelvin Yen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| | - Brendan Miller
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hiroshi Kumagai
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Ana Silverstein
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
18
|
Soukar J, Peppas NA, Gaharwar AK. Organelle-Targeting Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411720. [PMID: 39806939 PMCID: PMC11831507 DOI: 10.1002/advs.202411720] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/05/2024] [Indexed: 01/16/2025]
Abstract
Organelles are specialized subunits within cells which carry out vital functions crucial to cellular survival and form a tightly regulated network. Dysfunctions in any of these organelles are linked to numerous diseases impacting virtually every organ system in the human body. Targeted delivery of therapeutics to specific organelles within the cell holds great promise for overcoming challenging diseases and improving treatment outcomes through the minimization of therapeutic dosage and off-target effects. Nanoparticles are versatile and effective tools for therapeutic delivery to specific organelles. Nanoparticles offer several advantageous characteristics, including a high surface area-to-volume ratio for efficient therapeutic loading and the ability to attach targeting moieties (tethers) that enhance delivery. The choice of nanoparticle shape, size, composition, surface properties, and targeting ligands depends on the desired target organelle and therapeutic effect. Various nanoparticle platforms have been explored for organelle targeting, such as liposomes, polymeric nanoparticles, dendrimers, and inorganic nanoparticles. In this review, current and emerging approaches to nanoparticle design are examined in the context of various diseases linked to organelle dysfunction. Specifically, advances in nanoparticle therapies targeting organelles such as the nucleus, mitochondria, lysosomes/endosomes, Golgi apparatus, and endoplasmic reticulum are comprehensively and critically discussed.
Collapse
Affiliation(s)
- John Soukar
- Interdisiplinary program in Genetics and GenomicsTexas A&M UniversityCollege StationTX77843USA
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Nicholas A. Peppas
- Department of Biomedical EngineeringUniversity of Texas at AustinAustinTX78712USA
- Institute of BiomaterialsDrug Delivery and Regenerative MedicineUniversity of Texas at AustinAustinTX78712USA
- Department of Chemical EngineeringUniversity of Texas at AustinAustinTX78712USA
- Department of Surgery and Perioperative CareDell Medical SchoolUniversity of Texas at AustinAustinTX78712USA
- Department of PediatricsDell Medical SchoolUniversity of Texas at AustinAustinTX78712USA
| | - Akhilesh K. Gaharwar
- Interdisiplinary program in Genetics and GenomicsTexas A&M UniversityCollege StationTX77843USA
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
19
|
Kobayashi H, Matsubara S, Yoshimoto C, Shigetomi H, Imanaka S. A Comprehensive Review of the Contribution of Mitochondrial DNA Mutations and Dysfunction in Polycystic Ovary Syndrome, Supported by Secondary Database Analysis. Int J Mol Sci 2025; 26:1172. [PMID: 39940939 PMCID: PMC11818232 DOI: 10.3390/ijms26031172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder affecting women of reproductive age characterized by a spectrum of clinical, metabolic, reproductive, and psychological abnormalities. This syndrome is associated with significant long-term health risks, necessitating elucidation of its pathophysiology, early diagnosis, and comprehensive management strategies. Several contributory factors in PCOS, including androgen excess and insulin resistance, collectively enhance oxidative stress, which subsequently leads to mitochondrial dysfunction. However, the precise mechanisms through which oxidative stress induces mitochondrial dysfunction remain incompletely understood. Comprehensive searches of electronic databases were conducted to identify relevant studies published up to 30 September 2024. Mitochondria, the primary sites of reactive oxygen species (ROS) generation, play critical roles in energy metabolism and cellular homeostasis. Oxidative stress can inflict damage on components, including lipids, proteins, and DNA. Damage to mitochondrial DNA (mtDNA), which lacks efficient repair mechanisms, may result in mutations that impair mitochondrial function. Dysfunctional mitochondrial activity further amplifies ROS production, thereby perpetuating oxidative stress. These disruptions are implicated in the complications associated with the syndrome. Advances in genetic analysis technologies, including next-generation sequencing, have identified point mutations and deletions in mtDNA, drawing significant attention to their association with oxidative stress. Emerging data from mtDNA mutation analyses challenge conventional paradigms and provide new insights into the role of oxidative stress in mitochondrial dysfunction. We are rethinking the pathogenesis of PCOS based on these database analyses. In conclusion, this review explores the intricate relationship between oxidative stress, mtDNA mutations, and mitochondrial dysfunction, offers an updated perspective on the pathophysiology of PCOS, and outlines directions for future research.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara 634-0813, Japan;
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
- Department of Medicine, Kei Oushin Clinic, 5-2-6 Naruo-cho, Nishinomiya 663-8184, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, 2-897-5 Shichijyonishi-machi, Nara 630-8581, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, 3-3-17 Kitatomigaoka-cho, Nara 634-0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara 634-0813, Japan;
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.M.); (C.Y.); (H.S.)
| |
Collapse
|
20
|
Beresford-Webb JA, McAllister CJ, Sleigh A, Walpert MJ, Holland AJ, Zaman SH. Mitochondrial Dysfunction Correlates with Brain Amyloid Binding, Memory, and Executive Function in Down Syndrome: Implications for Alzheimer's Disease in Down Syndrome. Brain Sci 2025; 15:130. [PMID: 40002463 PMCID: PMC11853603 DOI: 10.3390/brainsci15020130] [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: 11/30/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Mitochondrial dysfunction is increasingly recognized as a central contributor to neurodegenerative diseases and age-related cognitive decline. Individuals with Down syndrome (DS) are at high risk of neurodegeneration due to Alzheimer's disease (AD). This study aims to explore the relationship between mitochondrial dysfunction, brain amyloid-beta (Aβ) deposition, and cognitive decline in this population. Methods: We investigated mitochondrial function, brain amyloid-beta burden, and cognitive performance in a pilot study of a cohort of 10 eligible adults with DS selected from a sample of 28 individuals with DS. Phosphorus-31 magnetic resonance spectroscopy (31P-MRS) was used to assess mitochondrial function in skeletal muscle using a post-exercise paradigm, while positron emission tomography using 11C-Pittsburgh compound B (PiB-PET) measured brain Aβ deposition. Cognitive performance was evaluated using the Cambridge Cognitive Examination adapted for individuals with Down syndrome (CAMCOG-DS) and executive function batteries. Results: Significant correlations were observed between slowed phosphocreatine (PCr) recovery in muscle and increased Aβ deposition in key brain regions, particularly the striatum. Cognitive performance inversely correlated with mitochondrial function, with pronounced deficits in memory and executive function tasks. Notably, an individual carrying the APOE-ε4 allele exhibited the poorest mitochondrial function, highest Aβ burden, and most severe cognitive impairment, suggesting a potential interaction between genetic risk and mitochondrial health. Conclusions: These findings highlight the role of mitochondrial dysfunction in DS-associated AD (DSAD) and its impact on cognition in adults. The results support targeting mitochondrial pathways as a potential therapeutic strategy to mitigate AD progression in DS populations. Further research with larger cohorts and longitudinal designs is needed to clarify causative mechanisms and develop effective interventions.
Collapse
Affiliation(s)
- Jessica A. Beresford-Webb
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Douglas House, Trumpington Road, Cambridge CB2 8AH, UK
| | - Catherine J. McAllister
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Douglas House, Trumpington Road, Cambridge CB2 8AH, UK
| | - Alison Sleigh
- Wolfson Brain Imaging Centre, University of Cambridge and NIHR Cambridge Clinical Research Facility, Cambridge University Hospitals NHS Foundation Trust, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Madeleine J. Walpert
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Douglas House, Trumpington Road, Cambridge CB2 8AH, UK
| | - Anthony J. Holland
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Douglas House, Trumpington Road, Cambridge CB2 8AH, UK
| | - Shahid H. Zaman
- Cambridge Intellectual and Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Douglas House, Trumpington Road, Cambridge CB2 8AH, UK
- Cambridgeshire & Peterborough Foundation NHS Trust, Douglas House, Trumpington Road, Cambridge CB2 8AH, UK
| |
Collapse
|
21
|
Xie L, Huang Y, Ma X, Ma X, Wang J, Gao T, Chen W. Effects of subclinical hypothyroidism during pregnancy on mtDNA methylation in the brain of rat offspring. BMC Neurosci 2025; 26:6. [PMID: 39856545 PMCID: PMC11762456 DOI: 10.1186/s12868-025-00930-2] [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: 08/14/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE This study aims to investigate the impact of subclinical hypothyroidism (SCH) during pregnancy on mitochondrial DNA (mtDNA) methylation in the brain tissues of rat offspring. MATERIALS AND METHODS Sixteen SD rats were randomly divided into two groups: control group (CON) and SCH group. BS-seq sequencing was used to analyze mtDNA methylation levels in the offspring's brain tissues; the 2,7-dichlorofluorescin diacetate (DCFH-DA) probe method was employed to detect reactive oxygen species (ROS) levels in brain tissues; electron microscopy was utilized to observe the mitochondrial structure in the hippocampal tissues of the offspring. RESULTS In the analysis of differentially methylated regions (DMRs), the mitochondrial chromosome in the SCH group exhibited 23 DMRs compared to the control group. ROS levels in the brain tissues of the SCH group were significantly higher than those in the control group (P < 0.05). The mitochondrial structure in the hippocampus of the SCH group was less intact compared to the CON group. CONCLUSION Subclinical hypothyroidism in pregnant rats may alter the mtDNA methylation pattern in the brains of their offspring, potentially affecting mitochondrial function and structure.
Collapse
Affiliation(s)
- Liangzhuo Xie
- Liaoning University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, P. R. China
| | - Yangling Huang
- Liaoning University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, P. R. China
| | - Xiande Ma
- Liaoning University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, P. R. China
| | - Xiaoqiu Ma
- Liaoning University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, P. R. China
| | - Jian Wang
- Experimental Animal Center of Liaoning, University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, P. R. China
| | - Tianshu Gao
- Department of Endocrine, Affiliated Hospital, Liaoning University of TCM, Shenyang City, Liaoning Province, P. R. China.
| | - Wei Chen
- Liaoning University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, P. R. China.
- The Second Affiliated Hospital of Liaoning, University of Traditional Chinese Medicine, Shenyang City, Liaoning Province, P. R. China.
| |
Collapse
|
22
|
Li-Harms X, Lu J, Fukuda Y, Lynch J, Sheth A, Pareek G, Kaminski MM, Ross HS, Wright CW, Smith AL, Wu H, Wang YD, Valentine M, Neale G, Vogel P, Pounds S, Schuetz JD, Ni M, Kundu M. Somatic mtDNA mutation burden shapes metabolic plasticity in leukemogenesis. SCIENCE ADVANCES 2025; 11:eads8489. [PMID: 39742470 PMCID: PMC11691655 DOI: 10.1126/sciadv.ads8489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
Abstract
The role of somatic mitochondrial DNA (mtDNA) mutations in leukemogenesis remains poorly characterized. To determine the impact of somatic mtDNA mutations on this process, we assessed the leukemogenic potential of hematopoietic progenitor cells (HPCs) from mtDNA mutator mice (Polg D257A) with or without NMyc overexpression. We observed a higher incidence of spontaneous leukemogenesis in recipients transplanted with heterozygous Polg HPCs and a lower incidence of NMyc-driven leukemia in those with homozygous Polg HPCs compared to controls. Although mtDNA mutations in heterozygous and homozygous HPCs caused similar baseline impairments in mitochondrial function, only heterozygous HPCs responded to and supported altered metabolic demands associated with NMyc overexpression. Homozygous HPCs showed altered glucose utilization with pyruvate dehydrogenase inhibition due to increased phosphorylation, exacerbated by NMyc overexpression. The impaired growth of NMyc-expressing homozygous HPCs was partially rescued by inhibiting pyruvate dehydrogenase kinase, highlighting a relationship between mtDNA mutation burden and metabolic plasticity in leukemogenesis.
Collapse
Affiliation(s)
- Xiujie Li-Harms
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jingjun Lu
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yu Fukuda
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - John Lynch
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Aditya Sheth
- Department of Pathology, Center of Excellence for Leukemia Studies, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Gautam Pareek
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Marcin M. Kaminski
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Hailey S. Ross
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Christopher W. Wright
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Amber L. Smith
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Huiyun Wu
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Marc Valentine
- Cytogenetics Shared Resource, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Peter Vogel
- Veterinary Pathology Core, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - John D. Schuetz
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Min Ni
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mondira Kundu
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
23
|
Jawla N, Kar R, Patil VS, Arimbasseri GA. Inherent metabolic preferences differentially regulate the sensitivity of Th1 and Th2 cells to ribosome-inhibiting antibiotics. Immunology 2025; 174:73-91. [PMID: 39263985 DOI: 10.1111/imm.13860] [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: 12/12/2023] [Accepted: 08/13/2024] [Indexed: 09/13/2024] Open
Abstract
Mitochondrial translation is essential to maintain mitochondrial function and energy production. Mutations in genes associated with mitochondrial translation cause several developmental disorders, and immune dysfunction is observed in many such patients. Besides genetic mutations, several antibiotics targeting bacterial ribosomes are well-established to inhibit mitochondrial translation. However, the effect of such antibiotics on different immune cells is not fully understood. Here, we addressed the differential effect of mitochondrial translation inhibition on different subsets of helper T cells (Th) of mice and humans. Inhibition of mitochondrial translation reduced the levels of mitochondrially encoded electron transport chain subunits without affecting their nuclear-encoded counterparts. As a result, mitochondrial oxygen consumption reduced dramatically, but mitochondrial mass was unaffected. Most importantly, we show that inhibition of mitochondrial translation induced apoptosis, specifically in Th2 cells. This increase in apoptosis was associated with higher expression of Bim and Puma, two activators of the intrinsic pathway of apoptosis. We propose that this difference in the sensitivity of Th1 and Th2 cells to mitochondrial translation inhibition reflects the intrinsic metabolic demands of these subtypes. Though Th1 and Th2 cells exhibit similar levels of oxidative phosphorylation, Th1 cells exhibit higher levels of aerobic glycolysis than Th2 cells. Moreover, Th1 cells are more sensitive to the inhibition of glycolysis, while higher concentrations of glycolysis inhibitor 2-deoxyglucose are required to induce cell death in the Th2 lineage. These observations reveal that selection of metabolic pathways for substrate utilization during differentiation of Th1 and Th2 lineages is a fundamental process conserved across species.
Collapse
Affiliation(s)
- Neha Jawla
- Molecular Genetics Laboratory, National Institute of Immunology, New Delhi, India
| | - Raunak Kar
- Immuno Genomics Laboratory, National Institute of Immunology, New Delhi, India
| | - Veena S Patil
- Immuno Genomics Laboratory, National Institute of Immunology, New Delhi, India
| | | |
Collapse
|
24
|
Bharadwaj A. A Review over Mitochondrial Diseases Due to mtDNA Mutations: Recent Advances and Remedial Aspects. Infect Disord Drug Targets 2025; 25:e18715265304029. [PMID: 39234902 DOI: 10.2174/0118715265304029240801092834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/01/2024] [Accepted: 06/13/2024] [Indexed: 09/06/2024]
Abstract
Mitochondria, also called 'powerhouse of the cell', is meant for energy generation in eukaryotic cells. This action is performed by mitochondria through the oxidative phosphorylation (OXPHOS) of the respiratory chain (RC). Based on the functioning of the cell, the number of mitochondria varies up to thousands in number. Mutations in the mitochondrial DNA (mtDNA) and/or nuclear DNA (nDNA) genes may lead to the generation of primary mitochondrial disease (PMD) that affects the structure and function of mitochondria. The diagnosis of such mitochondrial diseases occurs in early childhood and it can lead to serious, fetal and multi-organ diseases. Understanding epigenetic events and changes in the pathway can help improve the effectiveness of treatment. However, there are several reasons lack of the disease symptoms (age, sign, symptoms, morbidity and lethality), restricted availability of preclinical models along with extensive phenotypes that hamper the development of efficient drugs. Despite the introduction of new treatments and the encouraging results of treatments and therapies, there is no effective cure for PMD. This article contains information about the changes associated with cytopathic diseases that make possible the analysis of various diseases by genetic techniques. Increasing our understanding of how mitochondrial DNA mutations affect mitochondrial metabolism and subsequently result in neurodegenerative disease will prove vital to the development of targeted therapies and treatments.
Collapse
Affiliation(s)
- Alok Bharadwaj
- Department of Biotechnology, GLA University, Mathura (U.P.), India
| |
Collapse
|
25
|
de Menezes ECS, Malik AN. Absolute Quantification of Cellular and Cell-Free Mitochondrial DNA Copy Number from Human Blood and Urinary Samples Using Real Time Quantitative PCR. Methods Mol Biol 2025; 2878:233-257. [PMID: 39546266 DOI: 10.1007/978-1-0716-4264-1_13] [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] [Indexed: 11/17/2024]
Abstract
Mitochondrial DNA copy number (mtDNA-CN) in human body fluids is widely used as a biomarker of mitochondrial dysfunction in common metabolic diseases. Here we describe protocols to measure cellular and/or cell free (cf)-mtDNA-CN in human peripheral blood and urine. Cellular mtDNA is located inside the mitochondria where it encodes key subunits of the respiratory complexes in mitochondria and is usually normalized with reference to the nuclear genome as the mitochondrial genome to nuclear genome ratio (Mt/N) in either whole blood, peripheral blood mononuclear cells (PBMCs), or whole urine. Cf -mtDNA is usually found outside of the mitochondria, often released following mitochondrial damage, can trigger inflammatory pathways, and is usually measured as mtDNA-CN per volume of the starting material. Here we describe how to (1) separate whole blood into PBMCs, plasma, and serum fractions and whole urine into urinary supernatant and pellet, (2) prepare DNA from each of these fractions, (3) prepare reference standards for absolute quantification, (4) carry out qPCR for either relative or absolute quantification from test samples, (5) analyze qPCR data, and (6) calculate the sample size to adequately power studies. The protocol presented here is suitable for high throughput use and can be modified to quantify mtDNA from other body fluids, human cells, and tissues.
Collapse
Affiliation(s)
- Eliane Caseiro Soares de Menezes
- Diabetes and Obesity Theme, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, School of Life Course Science, King's College London, London, UK
| | - Afshan Navid Malik
- Diabetes and Obesity Theme, School of Cardiovascular Medicine and Metabolic Sciences, Faculty of Life Sciences and Medicine , King's College London , London, UK.
| |
Collapse
|
26
|
Du T, Su H, Cao D, Meng Q, Zhang M, Liu Z, Li H. Mitochondria-targeted antioxidant mitoquinone mitigates vitrification-induced damage in mouse ovarian tissue by maintaining mitochondrial homeostasis via the p38 MAPK pathway. Eur J Med Res 2024; 29:593. [PMID: 39696534 DOI: 10.1186/s40001-024-02181-z] [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: 09/12/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE Ovarian tissue cryopreservation has become a promising alternative for fertility preservation in cancer patients, allowing ovarian tissue to be stored for future autotransplantation. Oxidative stress damage occurring during the cryopreservation process may impact tissue quality and function. This study aims to investigate the protective effects and potential mechanisms of Mitoquinone (MitoQ), a mitochondria-targeted derivative of the antioxidant ubiquinone, during the vitrification of ovarian tissue in mice. METHODS KGN cells were treated with various concentrations (0.1, 1, 10, and 50 μM) of MitoQ to determine the optimal concentration. Female ICR mice were divided into three groups: control, conventional vitrification, and MitoQ-supplemented vitrification. Ovarian samples were cryopreserved, thawed, and assessed for tissue morphology using Hematoxylin and Eosin (H&E) staining, and mitochondrial changes using immunofluorescence, transmission electron microscopy, and Western blot analysis. RNA sequencing (RNA-seq) was employed to explore potential protective mechanisms. Autotransplantation experiments were conducted, and the long-term effects of MitoQ on ovarian function were evaluated by counting follicle numbers through H&E staining and measuring serum estradiol and AMH levels using ELISA. RESULTS MitoQ at 1 μM was found to be the optimal concentration for maintaining follicular morphology after vitrification. It effectively reduced mitochondrial oxidative damage, preserved mitochondrial morphology, and regulated the expression of mitochondrial dynamics proteins (Drp1 and Mfn2). RNA-seq and Western blot analyses revealed that MitoQ inhibited the p38 MAPK pathway, thereby reducing apoptosis. Additionally, autotransplantation experiments showed that MitoQ treatment significantly increased follicle counts, estradiol (E2), and anti-Müllerian hormone (AMH) levels compared to conventional vitrification. CONCLUSIONS MitoQ effectively mitigates vitrification-induced oxidative damage, maintains mitochondrial homeostasis, and preserves both follicular reserve and endocrine function. These findings suggest that MitoQ is a valuable adjunct in ovarian tissue cryopreservation and could significantly improve fertility preservation outcomes for cancer patients.
Collapse
Affiliation(s)
- Tianqi Du
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
| | - Han Su
- Key Laboratory of Reproductive Medicine and Offspring Health, Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhouing, China
- Obstetrics and Gynecology Department, BENQ Medical Center, Nanjing, China
| | - Dan Cao
- Department of Pathology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhouing, China
| | - Qingxia Meng
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
| | - Ming Zhang
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
- Key Laboratory of Reproductive Medicine and Offspring Health, Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhouing, China
| | - Zhenxing Liu
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
| | - Hong Li
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China.
| |
Collapse
|
27
|
Liu J, Yan Y, Zhang Y, Pan X, Xia H, Zhou J, Wan F, Huang X, Zhang W, Zhang Q, Chen B, Wang Y. Lysosome-Mitochondria Cascade Targeting Nanoparticle Drives Robust Pyroptosis for Cancer Immunotherapy. J Am Chem Soc 2024; 146:34568-34582. [PMID: 39639594 DOI: 10.1021/jacs.4c12264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The subcellular distribution of cargoes plays a crucial role in determining cell fate and therapeutic efficacy. However, achieving the precise delivery of therapeutics to specific intracellular targets remains a significant challenge. Here, we present a trimodular and acid/enzyme-gated nanoplatform (TAEN) that undergoes disassembly within acidic endosomes and then is cleaved by lysosomal cathepsin B to facilitate efficient and targeted transport of released cargoes into mitochondria compartments. By utilizing this nanovehicle, we successfully achieve selective sorting of photosensitizer molecules into mitochondria with a colocalization coefficient of up to 0.98, leading to the generation of reactive oxygen species stress specifically within the mitochondria for potent pyroptosis-based cancer therapy. The induction of mitochondrial stress triggers the intrinsic apoptotic pathway as well as caspase-3/gasdermin-E (GSDME) cascade, resulting in an enhanced cancer cell killing efficacy by nearly 2 orders of magnitude as compared to lysosomal stress. Furthermore, due to its superior capability to stimulate both innate and adaptive immune responses, our mitochondria-sorted nanophotosensitizer exhibits robust antitumor immune efficacy in multiple tumor-bearing mice models. This study not only provides insights into engineering nanomedicines for subcellular targeted delivery but also offers a valuable toolkit for advanced research in the field of nanobiology at subcellular resolution.
Collapse
Affiliation(s)
- Jianxiong Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yue Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yimeng Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xingquan Pan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiayi Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Fangjie Wan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinyu Huang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Weiwei Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Binlong Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Chemical Biology Center, Peking University, Beijing 100191, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo 315832, China
| |
Collapse
|
28
|
Haynes J, Joshi A, Larue RC, Eisenmann ED, Govindarajan R. Nucleoside Reverse Transcriptase Inhibitor (NRTI)-Induced Neuropathy and Mitochondrial Toxicity: Limitations of the Poly-γ Hypothesis and the Potential Roles of Autophagy and Drug Transport. Pharmaceutics 2024; 16:1592. [PMID: 39771570 PMCID: PMC11677988 DOI: 10.3390/pharmaceutics16121592] [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: 10/28/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are the backbone of highly active antiretroviral therapy (HAART)-the current standard of care for treating human immunodeficiency virus (HIV) infection. Despite their efficacy, NRTIs cause numerous treatment-limiting adverse effects, including a distinct peripheral neuropathy, called antiretroviral toxic neuropathy (ATN). ATN primarily affects the extremities with shock-like tingling pain, a pins-and-needles prickling sensation, and numbness. Despite its negative impact on patient quality of life, ATN remains poorly understood, which limits treatment options and potential interventions for people living with HIV (PLWH). Elucidating the underlying pathophysiology of NRTI-induced ATN will facilitate the development of effective treatment strategies and improved patient outcomes. In this article, we will comprehensively review ATN in the setting of NRTI treatment for HIV infection.
Collapse
Affiliation(s)
- John Haynes
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (J.H.); (A.J.); (E.D.E.)
| | - Arnav Joshi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (J.H.); (A.J.); (E.D.E.)
| | - Ross C. Larue
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Eric D. Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (J.H.); (A.J.); (E.D.E.)
- Translational Therapeutics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (J.H.); (A.J.); (E.D.E.)
- Translational Therapeutics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
29
|
Nie Q, Zhang W, Lin S, Huang M, Li Y, Qiu Y, Li J, Chen X, Wang Y, Tong X, Wu J, He P, Cai Q, Chen L, Chen M, Guo W, Lin Y, Yu L, Hou J, Cai W, Chen H, Wang C, Fu F. Identification of sequence polymorphism in the D-loop region of mitochondrial DNA as a risk factor for breast cancer. Cancer Sci 2024; 115:4064-4073. [PMID: 39401980 PMCID: PMC11611757 DOI: 10.1111/cas.16353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/23/2024] [Accepted: 09/08/2024] [Indexed: 12/06/2024] Open
Abstract
Mitochondrial DNA (mtDNA) variations affect the efficiency of the electron transport chain and production of reactive oxygen species, contributing to carcinogenesis. The D-loop region of mtDNA has emerged as a variation hotspot region in human neoplasia; however, the potential contribution of these variations in breast cancer risk prediction remains unknown. We investigated the relationship between germline single nucleotide polymorphisms (SNPs) in the entire D-loop region and breast cancer risk in Chinese women. Peripheral blood-isolated mtDNA from 2329 patients with breast cancer and 2328 cancer-free controls was examined for SNPs. In the combined cohort, we used traditional risk factors, susceptibility germline polymorphisms, and logistic regression analysis to evaluate the predictive value of susceptibility variants for breast cancer risk. We calculated the area under the receiver operating characteristic curve (AUC) as a measure. We also measured the content of 8-hydroxy-2'-deoxyguanosine (8-OHdG). Individual polymorphisms SNP573 were significantly associated with breast cancer risk in both the discovery and validation cohorts. In the combined cohort, the AUC of the traditional risk factors was 64.3%; after adding susceptibility variants, the AUC was 64.9% (DeLong test, p = 0.007). 8-OHdG levels were significantly higher in patients with breast cancer than in controls and higher in individuals with SNP573 than in those negative for this variation. Overall, oxidative stress might be associated with the risk of breast cancer, and SNP573 might be associated with oxidative stress. Our results indicate the risk potential of polymorphisms in the D-loop region in breast cancer in Southern China.
Collapse
|
30
|
Rossi T, Iorio E, Chirico M, Pisanu ME, Amodio N, Cantafio MEG, Perrotta I, Colciaghi F, Fiorillo M, Gianferrari A, Puccio N, Neri A, Ciarrocchi A, Pistoni M. BET inhibitors (BETi) influence oxidative phosphorylation metabolism by affecting mitochondrial dynamics leading to alterations in apoptotic pathways in triple-negative breast cancer (TNBC) cells. Cell Prolif 2024; 57:e13730. [PMID: 39223828 PMCID: PMC11628750 DOI: 10.1111/cpr.13730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Repressing BET proteins' function using bromodomain inhibitors (BETi) has been shown to elicit antitumor effects by regulating the transcription of genes downstream of BRD4. We previously showed that BETi promoted cell death of triple-negative breast cancer (TNBC) cells. Here, we proved that BETi induce altered mitochondrial dynamics fitness in TNBC cells falling in cell death. We demonstrated that BETi treatment downregulated the expression of BCL-2, and proteins involved in mitochondrial fission and increased fused mitochondria. Impaired mitochondrial fission affected oxidative phosphorylation (OXPHOS) inducing the expression of OXPHOS-related genes, SDHa and ATP5a, and increased cell death. Consistently, the amount of mitochondrial DNA and mitochondrial membrane potential (∆Ψm) increased in BETi-treated cells compared to control cells. Lastly, BETi in combination with Metformin reduced cell growth. Our results indicate that mitochondrial dynamics and OXPHOS metabolism support breast cancer proliferation and represent novel BETi downstream targets in TNBC cells.
Collapse
Affiliation(s)
- Teresa Rossi
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Egidio Iorio
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Mattea Chirico
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Maria Elena Pisanu
- High Resolution NMR UnitCore Facilities, Istituto Superiore di SanitàRomeItaly
| | - Nicola Amodio
- Department of Experimental and Clinical MedicineUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | | | - Ida Perrotta
- Department of Biology, Ecology and Earth SciencesCentre for Microscopy and Microanalysis (CM2), University of CalabriaCosenzaItaly
| | | | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional SciencesUniversity of CalabriaRendeItaly
| | - Alessia Gianferrari
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Noemi Puccio
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Antonino Neri
- Scientific DirectorateAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Alessia Ciarrocchi
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| | - Mariaelena Pistoni
- Laboratory of Translational ResearchAUSL‐IRCCS di Reggio EmiliaReggio EmilaItaly
| |
Collapse
|
31
|
Han J, Xu W, Yu H, Han Y, Zhu M. Structural and evolutionary analyses of the mitochondrial genome of Spuriopimpinella brachycarpa. FRONTIERS IN PLANT SCIENCE 2024; 15:1492723. [PMID: 39659412 PMCID: PMC11628310 DOI: 10.3389/fpls.2024.1492723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024]
Abstract
Introduction Spuriopimpinella brachycarpa (Kom.) Kitag., a member of the Apiaceae family, is a perennial aromatic herb native to Northeast Asia with applications in culinary and traditional medicine. Despite its significance, most studies on S. brachycarpa have primarily focused on its phytochemical properties, with limited insights into its molecular and genomic characteristics. Methods This study presents the sequencing and assembly of the mitochondrial genome (mitogenome) of S. brachycarpa using second- and third-generation high-throughput sequencing technologies. Comprehensive analyses were performed on its structural organization, RNA editing sites, relative synonymous codon usage (RSCU), and repeat sequences. Comparative analyses with closely related species were also conducted. Results The mitogenome exhibited a multi-branched structure, with a total length of 523,512 bp and a GC content of 43.37%. Annotation revealed 30 unique protein-coding genes, 21 tRNA genes, and three rRNA genes. Comparative analysis indicated that the S. brachycarpa mitogenome contains structural variations but shares collinear features with other Apiaceae species. We identified 618 potential RNA editing sites involving C-to-U conversions and discovered 59 homologous fragments between the mitogenome and plastome, comprising 8.13% of the mitogenome. Discussion These results enrich the genomic database of Apiaceae, providing valuable insights into the evolutionary relationships and genetic diversity within the family.
Collapse
Affiliation(s)
- Jun Han
- Chinese Medicine Research Institute of Beijing Tcmages Pharmaceutical Co., Ltd., Beijing, China
| | - Wenbo Xu
- Chinese Medicine Research Institute of Beijing Tcmages Pharmaceutical Co., Ltd., Beijing, China
| | - Huanxi Yu
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment of the People’s Republic of China, Nanjing, China
| | - Yun Han
- College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Ming Zhu
- College of Life Sciences, South China Agricultural University, Guangzhou, China
| |
Collapse
|
32
|
Wu X, Wang K, Chen H, Cao B, Wang Y, Wang Z, Dai C, Yao M, Ji X, Jiang X, Zhang W, Pan Z, Xue D. Hypoxia-induced mitochondrial fission regulates the fate of bone marrow mesenchymal stem cells by maintaining HIF1α stabilization. Free Radic Biol Med 2024; 225:127-144. [PMID: 39366470 DOI: 10.1016/j.freeradbiomed.2024.10.256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/19/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
For mesenchymal stem cells derived from bone marrow, a controlled reduction in ambient oxygen concentration has been recognized as a facilitator of osteogenic differentiation and the formation of calcium nodules. However, the specific molecular mechanisms underlying this phenotype remain unclear. The aim of this study was to elucidate the impact of hypoxia on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and to explore the involvement of mitophagy and the regulation of mitochondrial dynamics mediated by the mitochondrial dynamic regulatory factor FUN14 domain-containing 1 (FUNDC1). Our findings suggest that FUNDC1 is required for promoting osteogenic differentiation in BMSCs under hypoxic conditions. However, this effect was not dependent on FUNDC1-mediated mitophagy but rather on FUNDC1-mediated regulation of mitochondrial fission. At the mechanistic level, FUNDC1 binds more DNM1L and less OPA1 under hypoxic conditions, leading to an upsurge in mitochondrial division. This heightened mitochondrial division culminates in the increased translocation of Parkin to mitochondria, diminishing its interactions with HIF1α in the cytoplasm and consequently facilitating HIF1α deubiquitination and stabilization. In summary, FUNDC1-regulated mitochondrial division in hypoxic culture emerges as a critical determinant for the translocation of Parkin to mitochondria, ultimately maintaining HIF1α stabilization and promoting osteogenic differentiation.
Collapse
Affiliation(s)
- Xiaoyong Wu
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Kanbin Wang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Hongyu Chen
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Binhao Cao
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Yibo Wang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhongxiang Wang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Chengxin Dai
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Minjun Yao
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Xiaoxiao Ji
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Xiaowen Jiang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Weijun Zhang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Zhijun Pan
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China.
| | - Deting Xue
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
33
|
Shen L, Zhang L, Jin J, Jin Z, Li Z, Wu L, Cheng K, Xu D, Liu H. The phototoxicity of sulfamethoxazole stress on pakchoi cabbage (Brassica rapa var. chinensis) seedlings: From the perspective of photoreaction and omics analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175391. [PMID: 39122040 DOI: 10.1016/j.scitotenv.2024.175391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
The increasing use of antibiotics has attracted widespread attention to their environmental risks. However, the phototoxicity of sulfonamide antibiotics to plants remain unclear. In this study, the mechanism of the effect of sulfamethoxazole on photosynthesis of pakchoi cabbage (Brassica rapa var. chinensis) was investigated. The results showed that sulfamethoxazole inhibited the growth of pakchoi cabbage and produced photosynthetic toxicity. The growth inhibition rates increased with concentration, the root and shoot weight were 76.02 % and 47.04 % of the control, respectively, with stay-greens phenomenon in 4 mg·L-1 sulfamethoxazole treatment. Chlorophyll precursors (protoporphyrin IX (Proto IX), Mg-proto IX, and protochlorophyllide (Pchlide), 5-aminolevulinic acid (ALA), and porphobilinogen (PBG)) were 1.38-, 1.26-, 1.12-, 1.71-, and 0.96-fold of the control, respectively; photosynthetic pigments (chlorophyll a, chlorophyll b, and carotenoids) were 1.26-, 1.39-, and 1.03-fold of the control, respectively. Respiration rate was 271.42 % of the control, whereas the net photosynthetic rate was 50.50 % of the control. The maximum photochemical quantum yield of PSII (Fv/Fm), the actual photosynthetic efficiency (Y(II)), the quantum yield of non-regulated energy dissipation (Y(NO)), the apparent electron transfer efficiency of PSII (ETR) under actual light intensity were affected, and chloroplast swelling was observed. Proteomic analysis showed that photosynthesis-related pathways were significantly up-regulated, biological processes such as light response, carbohydrates, and reactive oxygen species were activated. Metabolomic analysis revealed that the tricarboxylic acid cycle (TCA cycle) and carbohydrate catabolism were stimulated significantly (p < 0.05), sugars and amino acids were increased to regulate and enhance the resilience of photosynthesis. While folate biosynthesis and ribosomal pathways were significantly down-regulated, the synthesis and translation processes of amino acids and nucleotides were inhibited.
Collapse
Affiliation(s)
- Luoqin Shen
- School of Environmental Science and Engineering, Key Laboratory of Solid Waste Treatment and Recycling of Zhejiang Province, International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang Province, China
| | - Liangyu Zhang
- School of Environmental Science and Engineering, Key Laboratory of Solid Waste Treatment and Recycling of Zhejiang Province, International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang Province, China
| | - Jiaojun Jin
- School of Environmental Science and Engineering, Key Laboratory of Solid Waste Treatment and Recycling of Zhejiang Province, International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang Province, China
| | - Ziting Jin
- School of Environmental Science and Engineering, Key Laboratory of Solid Waste Treatment and Recycling of Zhejiang Province, International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang Province, China
| | - Zhiheng Li
- School of Environmental Science and Engineering, Key Laboratory of Solid Waste Treatment and Recycling of Zhejiang Province, International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang Province, China
| | - Lidan Wu
- School of Environmental Science and Engineering, Key Laboratory of Solid Waste Treatment and Recycling of Zhejiang Province, International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang Province, China
| | - Kaiming Cheng
- School of Statistics and Mathematics, Collaborative Innovation Center of Statistical Data Engineering, Technology & Application, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Dongmei Xu
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, College of Biological and Environmental Engineering, Zhejiang Shuren University, Hangzhou 310015, Zhejiang Province, China
| | - Huijun Liu
- School of Environmental Science and Engineering, Key Laboratory of Solid Waste Treatment and Recycling of Zhejiang Province, International Science and Technology Cooperation Platform for Low-Carbon Recycling of Waste and Green Development, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang Province, China.
| |
Collapse
|
34
|
Wang R, Bao F, Lu M, Jia X, Xiao J, Wu Y, Zhang Q, Liu X. MSC-mediated mitochondrial transfer restores mitochondrial DNA and function in neural progenitor cells of Leber's hereditary optic neuropathy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2511-2519. [PMID: 39134891 DOI: 10.1007/s11427-024-2647-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/21/2024] [Indexed: 10/22/2024]
Abstract
Leber's hereditary optic neuropathy (LHON) is a debilitating mitochondrial disease associated with mutations in mitochondrial DNA (mtDNA). Unfortunately, the available treatment options for LHON patients are limited due to challenges in mitochondrial replacement. In our study, we reprogramming LHON urine cells into induced pluripotent stem cells (iPSCs) and differentiating them into neural progenitor cells (NPCs) and neurons for disease modeling. Our research revealed that LHON neurons exhibited significantly higher levels of mtDNA mutations and reduced mitochondrial function, confirming the disease phenotype. However, through co-culturing LHON iPSC-derived NPCs with mesenchymal stem cells (MSCs), we observed a remarkable rescue of mutant mtDNA and a significant improvement in mitochondrial metabolic function in LHON neurons. These findings suggest that co-culturing with MSCs can enhance mitochondrial function in LHON NPCs, even after their differentiation into neurons. This discovery holds promise as a potential therapeutic strategy for LHON patients.
Collapse
Affiliation(s)
- Rui Wang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, 99077, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Feixiang Bao
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Manjiao Lu
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xiaoyun Jia
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jiahui Xiao
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yi Wu
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Qingjiong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Xingguo Liu
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, 510530, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, 99077, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
35
|
Alves JL, Reis PM, Quinta-Ferreira RM, Quinta-Ferreira ME, Matias CM. Changes in reactive oxygen species and autofluorescence under hypoxia at the hippocampal CA3 area: Role of calcium and zinc influxes. Neurochem Int 2024; 180:105882. [PMID: 39413928 DOI: 10.1016/j.neuint.2024.105882] [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: 05/09/2024] [Revised: 09/12/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Reactive oxygen species (ROS) have an important role in cellular biology, being involved, in a way that depends on their levels, in cell signaling processes or in oxidative stress, probably associated with neurodegenerative and other diseases. Most of the studies about ROS formation were performed in ischemic conditions, and thus, there is limited knowledge about ROS formation in less severe hypoxic conditions. This study investigates neuronal ROS generation and autofluorescence changes in hypoxic conditions, focusing on the involvement of calcium and zinc. Using hippocampal slices from Wistar rats, ROS production was monitored by the permeant fluorescent indicator H2DCFDA under different oxygenation levels. Moderate hypoxia (40% O2) led to a small ROS increase, while severe hypoxia (0% O2) showed a more pronounced rise. KCl-induced depolarization significantly enhanced ROS formation, particularly under severe hypoxia. Inhibition of NMDA receptors reduced ROS generation without affecting autofluorescence, while chelation of zinc ions decreased ROS production and increased flavin adenine dinucleotide (FAD) autofluorescence. These findings suggest that, in hypoxic conditions, ROS formation is mediated by calcium entry through NMDA receptors and also by zinc influxes. Thus, these ions play a crucial role in oxidative stress, which may be related with neurodegenerative diseases associated with ROS dysregulation.
Collapse
Affiliation(s)
- João L Alves
- Department of Life Sciences, University of Coimbra, Portugal; CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Patrícia M Reis
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - Rosa M Quinta-Ferreira
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - M Emília Quinta-Ferreira
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, University of Coimbra, Portugal
| | - Carlos M Matias
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, UTAD, Vila Real, Portugal.
| |
Collapse
|
36
|
Indo HP, Chatatikun M, Nakanishi I, Matsumoto KI, Imai M, Kawakami F, Kubo M, Abe H, Ichikawa H, Yonei Y, Beppu HJ, Minamiyama Y, Kanekura T, Ichikawa T, Phongphithakchai A, Udomwech L, Sukati S, Charong N, Somsak V, Tangpong J, Nomura S, Majima HJ. The Roles of Mitochondria in Human Being's Life and Aging. Biomolecules 2024; 14:1317. [PMID: 39456251 PMCID: PMC11506671 DOI: 10.3390/biom14101317] [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: 08/27/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
The universe began 13.8 billion years ago, and Earth was born 4.6 billion years ago. Early traces of life were found as soon as 4.1 billion years ago; then, ~200,000 years ago, the human being was born. The evolution of life on earth was to become individual rather than cellular life. The birth of mitochondria made this possible to be the individual life. Since then, individuals have had a limited time of life. It was 1.4 billion years ago that a bacterial cell began living inside an archaeal host cell, a form of endosymbiosis that is the development of eukaryotic cells, which contain a nucleus and other membrane-bound compartments. The bacterium started to provide its host cell with additional energy, and the interaction eventually resulted in a eukaryotic cell, with both archaeal (the host cell) and bacterial (mitochondrial) origins still having genomes. The cells survived high concentrations of oxygen producing more energy inside the cell. Further, the roles of mitochondria in human being's life and aging will be discussed.
Collapse
Affiliation(s)
- Hiroko P. Indo
- Department of Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City 890-8544, Japan; (H.P.I.)
- Amanogawa Galactic Astronomy Research Center (AGARC), Kagoshima University Graduate School of Sciences and Engineering, 1-21-40 Korimoto, Kagoshima 890-0065, Japan
| | - Moragot Chatatikun
- School of Allied Health Sciences, Walailak University, Thasala 80161, Thailand; (M.C.); (S.S.); (N.C.); (V.S.); (J.T.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Thasala Nakhon Si Thammarat 80160, Thailand
| | - Ikuo Nakanishi
- Quantum RedOx Chemistry Team, Quantum Life Spin Group, Institute for Quantum Life Science (iQLS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan;
| | - Ken-ichiro Matsumoto
- Quantitative RedOx Sensing Group, Department of Radiation Regulatory Science Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Motoki Imai
- Department of Molecular Diagnostics, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
- Department of Applied Tumor Pathology, Graduate School of Medical Sciences, Kitasato University, Sagamihara 252-0374, Japan
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
| | - Fumitaka Kawakami
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
- Department of Health Administration, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
| | - Makoto Kubo
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
- Department of Environmental Microbiology, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
| | - Hiroshi Abe
- Department of Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima City 890-8544, Japan; (H.P.I.)
| | - Hiroshi Ichikawa
- Department of Medical Life Systems, Graduate School of Life and Medical Sciences, Doshishia University, Kyoto 610-0394, Japan
| | - Yoshikazu Yonei
- Anti-Aging Medical Research Center and Glycation Stress Research Center, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto 610-0394, Japan
| | - Hisashi J. Beppu
- Dr. Beppu’s Oral Health Care & Anti-Aging Clinic, Chuo-ku, Tokyo 103-0027, Japan
| | - Yukiko Minamiyama
- Food Hygiene and Environmental Health Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto 606-8522, Japan
| | - Takuro Kanekura
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Takafumi Ichikawa
- Regenerative Medicine and Cell Design Research Facility, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Sagamihara 252-0373, Japan
| | - Atthaphong Phongphithakchai
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Lunla Udomwech
- School of Medicine, Walailak University, Thasala 80161, Thailand
| | - Suriyan Sukati
- School of Allied Health Sciences, Walailak University, Thasala 80161, Thailand; (M.C.); (S.S.); (N.C.); (V.S.); (J.T.)
- Hematology and Transfusion Science Research Center (HTSRC), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Nurdina Charong
- School of Allied Health Sciences, Walailak University, Thasala 80161, Thailand; (M.C.); (S.S.); (N.C.); (V.S.); (J.T.)
- Hematology and Transfusion Science Research Center (HTSRC), School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Voravuth Somsak
- School of Allied Health Sciences, Walailak University, Thasala 80161, Thailand; (M.C.); (S.S.); (N.C.); (V.S.); (J.T.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Thasala Nakhon Si Thammarat 80160, Thailand
| | - Jitbanjong Tangpong
- School of Allied Health Sciences, Walailak University, Thasala 80161, Thailand; (M.C.); (S.S.); (N.C.); (V.S.); (J.T.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Thasala Nakhon Si Thammarat 80160, Thailand
| | - Sachiyo Nomura
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan;
- Isotope Science Center, The University of Tokyo, 2-22-16 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
- Department of Gastrointestinal Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hideyuki J. Majima
- School of Allied Health Sciences, Walailak University, Thasala 80161, Thailand; (M.C.); (S.S.); (N.C.); (V.S.); (J.T.)
- Research Excellence Center for Innovation and Health Products (RECIHP), School of Allied Health Sciences, Walailak University, Thasala Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
37
|
Ishikawa K, Miyata D, Hattori S, Tani H, Kuriyama T, Wei FY, Miyakawa T, Nakada K. Accumulation of mitochondrial DNA with a point mutation in tRNA Leu(UUR) gene induces brain dysfunction in mice. Pharmacol Res 2024; 208:107374. [PMID: 39197713 DOI: 10.1016/j.phrs.2024.107374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Brain functions are mediated via the complex interplay between several complex factors, and hence, identifying the underlying cause of an abnormality within a certain brain region can be challenging. In mitochondrial disease, abnormalities in brain function are thought to be attributed to accumulation of mitochondrial DNA (mtDNA) with pathogenic mutations; however, only few previous studies have directly demonstrated that accumulation of mutant mtDNA induced abnormalities in brain function. Herein, we examined the effects of mtDNA mutations on brain function via behavioral analyses using a mouse model with an A2748G point mutation in mtDNA tRNALeu(UUR). Our results revealed that mice with a high percentage of mutant mtDNA showed a characteristic trend toward reduced prepulse inhibition and memory-dependent test performance, similar to that observed in psychiatric disorders, such as schizophrenia; however, muscle strength and motor coordination were not markedly affected. Upon examining the hippocampus and frontal lobes of the brain, mitochondrial morphology was abnormal, and the brain weight was slightly reduced. These results indicate that the predominant accumulation of a point mutation in the tRNALeu(UUR) gene may affect brain functions, particularly the coordination of sensory and motor functions and memory processes. These abnormalities probably caused by both direct effects of accumulation of the mutant mtDNA in neuronal cells and indirect effects via changes of systemic extracellular environments. Overall, these findings will lead to a better understanding of the pathogenic mechanism underlying this complex disease and facilitate the development of optimal treatment methods.
Collapse
Affiliation(s)
- Kaori Ishikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, Japan.
| | - Daiki Miyata
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Japan
| | - Satoko Hattori
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Japan; Research Creation Support Center, Aichi Medical University, Japan
| | - Haruna Tani
- Department of Modomics Biology and Medicine, Institute of Development, Aging and Cancer, Tohoku University, Japan
| | - Takayoshi Kuriyama
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Japan
| | - Fan-Yan Wei
- Department of Modomics Biology and Medicine, Institute of Development, Aging and Cancer, Tohoku University, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Japan
| | - Kazuto Nakada
- Institute of Life and Environmental Sciences, University of Tsukuba, Japan.
| |
Collapse
|
38
|
Roheel A, Khan A, Anwar F, Ullah H, Rehman AU, Ullah N, Akhtar MF, Khan MI, Yaseen N. Evaluation of anti-tumor activity of molybdenum disulfide nanoflowers per se and in combination with berberine against mammary gland cancer in rats. JOURNAL OF NANOPARTICLE RESEARCH 2024; 26:240. [DOI: 10.1007/s11051-024-06153-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/04/2024] [Indexed: 01/28/2025]
|
39
|
Tamvaka N, Heckman MG, Johnson PW, Soto-Beasley AI, Walton RL, Koga S, Uitti RJ, Parfitt F, Graff-Radford MR, Wszolek ZK, Graff-Radford N, Valentino RR, Ross OA. Associations of mitochondrial genomic variation with successful neurological aging. Mitochondrion 2024; 78:101948. [PMID: 39179138 DOI: 10.1016/j.mito.2024.101948] [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: 03/01/2024] [Revised: 07/25/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
Mitochondrial health is an integral factor in aging, with mitochondrial dysfunction known to increase with age and contribute to the development of age-related neurodegenerative disorders. Additionally, the mitochondrial genome (mtDNA) has been shown to acquire potentially damaging somatic variation as part of the aging process, while mtDNA single nucleotide polymorphism (SNPs) have been shown to be both protective and detrimental for various neurodegenerative diseases. Yet, little is known about the involvement of mtDNA variation in longevity and successful neurological aging. In this study, we examined the association of mtDNA SNPs, in the form of mitochondrial haplogroups, with successful neurological aging in 1,405 unrelated neurologically healthy subjects. Although not quite significant after correcting for multiple testing (P < 0.0017 considered as significant), we detected a nominally significant association between the I haplogroup (N = 45, 3.2 %) and a younger age (β: -5.00, P = 0.006), indicating that this haplogroup is observed less frequently in older neurologically healthy individuals and may be associated with decreased survival. Replication of this finding in independent neurologically healthy cohorts will be imperative for shaping our understanding of the biological processes underlying healthy neurological aging.
Collapse
Affiliation(s)
- Nicole Tamvaka
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA
| | - Michael G Heckman
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Patrick W Johnson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Ronald L Walton
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Francine Parfitt
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | | | | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA; Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Biology, University of North Florida, Jacksonville, FL 32224, USA; Department of Medicine, University College Dublin, Dublin, Ireland.
| |
Collapse
|
40
|
Singh D. Beyond the membrane: Exploring non-viral methods for mitochondrial gene delivery. Mitochondrion 2024; 78:101922. [PMID: 38897397 DOI: 10.1016/j.mito.2024.101922] [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: 04/10/2024] [Revised: 05/22/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Mitochondrial disorders, stemming from mutations in mitochondrial DNA (mtDNA), present a significant therapeutic challenge due to their complex pathophysiology and broad spectrum of clinical manifestations. Traditional gene therapy approaches, primarily reliant on viral vectors, face obstacles such as potential immunogenicity, insertional mutagenesis, and the specificity of targeting mtDNA. This review delves into non-viral methods for mitochondrial gene delivery, emerging as a promising alternative to overcome these limitations. Focusing on lipid-based nanoparticles, polymer-based vectors, and mitochondrial-targeted peptides, the mechanisms of action, advantages, and current applications in treating mitochondrial diseases was well elucidated. Non-viral vectors offer several benefits, including reduced immunogenicity, enhanced safety profiles, and the flexibility to carry a wide range of genetic material. We examine case studies where these methods have been applied, highlighting their potential in correcting pathogenic mtDNA mutations and mitigating disease phenotypes. Despite their promise, challenges such as delivery efficiency, specificity, and long-term expression stability persist. The review underscores the need for ongoing research to refine these delivery systems carry a wide range of genetic material. We examine case studies where these methods settings. As we advance our understanding of mitochondrial biology and gene delivery technologies, non-viral methods hold the potential to revolutionize the treatment of mitochondrial disorders, offering hope for therapies that can precisely target and correct the underlying genetic defects.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali 140413, India; University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India.
| |
Collapse
|
41
|
Natarajan N, Florentin J, Johny E, Xiao H, O'Neil SP, Lei L, Shen J, Ohayon L, Johnson AR, Rao K, Li X, Zhao Y, Zhang Y, Tavakoli S, Shiva S, Das J, Dutta P. Aberrant mitochondrial DNA synthesis in macrophages exacerbates inflammation and atherosclerosis. Nat Commun 2024; 15:7337. [PMID: 39187565 PMCID: PMC11347661 DOI: 10.1038/s41467-024-51780-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
There is a large body of evidence that cellular metabolism governs inflammation, and that inflammation contributes to the progression of atherosclerosis. However, whether mitochondrial DNA synthesis affects macrophage function and atherosclerosis pathology is not fully understood. Here we show, by transcriptomic analyzes of plaque macrophages, spatial single cell transcriptomics of atherosclerotic plaques, and functional experiments, that mitochondrial DNA (mtDNA) synthesis in atherosclerotic plaque macrophages are triggered by vascular cell adhesion molecule 1 (VCAM-1) under inflammatory conditions in both humans and mice. Mechanistically, VCAM-1 activates C/EBPα, which binds to the promoters of key mitochondrial biogenesis genes - Cmpk2 and Pgc1a. Increased CMPK2 and PGC-1α expression triggers mtDNA synthesis, which activates STING-mediated inflammation. Consistently, atherosclerosis and inflammation are less severe in Apoe-/- mice lacking Vcam1 in macrophages. Downregulation of macrophage-specific VCAM-1 in vivo leads to decreased expression of LYZ1 and FCOR, involved in STING signalling. Finally, VCAM-1 expression in human carotid plaque macrophages correlates with necrotic core area, mitochondrial volume, and oxidative damage to DNA. Collectively, our study highlights the importance of macrophage VCAM-1 in inflammation and atherogenesis pathology and proposes a self-acerbating pathway involving increased mtDNA synthesis.
Collapse
Affiliation(s)
- Niranjana Natarajan
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jonathan Florentin
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Ebin Johny
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Hanxi Xiao
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Joint CMU-Pitt PhD program in Computational Biology, Pittsburgh, PA, USA
| | - Scott Patrick O'Neil
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Liqun Lei
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Jixing Shen
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Lee Ohayon
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Aaron R Johnson
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Krithika Rao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Xiaoyun Li
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yanwu Zhao
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Yingze Zhang
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sina Tavakoli
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
- University of Pittsburgh School of Medicine Department of Pharmacology & Chemical Biology, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
42
|
Motegi M, Sakurai Y, Mio Y, Ohashi T. Cochlear Implantation for Isoleucyl-tRNA Synthetase Mutation-Associated Mitochondrial Disease: A Case Report. Cureus 2024; 16:e67760. [PMID: 39323698 PMCID: PMC11422515 DOI: 10.7759/cureus.67760] [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] [Accepted: 08/25/2024] [Indexed: 09/27/2024] Open
Abstract
Biallelic missense mutations in the nuclear-encoded, cytosolic isoleucyl-tRNA synthetase (IARS) gene are associated with a rare and complex multisystemic phenotype, including growth retardation, intellectual disability, muscular hypotonia, diabetes mellitus, and deafness. These mutations impact the cytosolic isoform of IARS, which plays a crucial role in protein synthesis. The pathogenesis involves mitochondrial dysfunction, despite IARS being primarily a cytosolic enzyme, potentially linking it to the observed clinical manifestations. The efficacy of cochlear implantation for deafness due to IARS mutations and the safety of general anesthesia in such patients remain unclear. This report describes a rare case of progressive sensorineural hearing loss caused by IARS mutation-associated mitochondrial disease, which was successfully treated with cochlear implantation. Additionally, we discuss the safety of general anesthesia in this patient. A Japanese woman with IARS mutation-associated mitochondrial disease was diagnosed with severe bilateral sensorineural hearing loss at five years of age and immediately received hearing aids. Her hearing progressively deteriorated to profound impairment, necessitating cochlear implantation at 26 years of age, which resulted in satisfactory hearing. Furthermore, no perioperative general anesthesia-related adverse events were reported. Our case demonstrates that cochlear implantation can effectively restore hearing. This suggests that sensorineural hearing loss caused by IARS deficiency is primarily due to cochlear dysfunction. This case demonstrated that hearing loss is a crucial feature of IARS mutation-associated mitochondrial disease, which can be mitigated by cochlear implantation. While general anesthesia can be safely administered, careful consideration of anesthetic choices, such as avoiding depolarizing muscle relaxants and prolonged use of propofol, is essential to prevent complications. In this case, general anesthesia was well tolerated without perioperative events, providing valuable insight into the potential safety of such procedures in similar patients. Nevertheless, further studies are needed to confirm these findings across a broader population.
Collapse
Affiliation(s)
- Masaomi Motegi
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine, Maebashi, JPN
| | - Yuika Sakurai
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Minato, JPN
| | - Yasushi Mio
- Department of Anesthesiology, The Jikei University School of Medicine, Minato, JPN
| | - Toya Ohashi
- Department of Human Health Science and Therapeutics, The Jikei University School of Nursing, Chofu, JPN
| |
Collapse
|
43
|
Yazdani M. Cellular and Molecular Responses to Mitochondrial DNA Deletions in Kearns-Sayre Syndrome: Some Underlying Mechanisms. Mol Neurobiol 2024; 61:5665-5679. [PMID: 38224444 DOI: 10.1007/s12035-024-03938-7] [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] [Indexed: 01/16/2024]
Abstract
Kearns-Sayre syndrome (KSS) is a rare multisystem mitochondrial disorder. It is caused by mitochondrial DNA (mtDNA) rearrangements, mostly large-scale deletions of 1.1-10 kb. These deletions primarily affect energy supply through impaired oxidative phosphorylation and reduced ATP production. This impairment gives rise to dysfunction of several tissues, in particular those with high energy demand like brain and muscles. Over the past decades, changes in respiratory chain complexes and energy metabolism have been emphasized, whereas little attention has been paid to other reports on ROS overproduction, protein synthesis inhibition, myelin vacuolation, demyelination, autophagy, apoptosis, and involvement of lipid raft and oligodendrocytes in KSS. Therefore, this paper draws attention towards these relatively underemphasized findings that might further clarify the pathologic cascades following deletions in the mtDNA.
Collapse
Affiliation(s)
- Mazyar Yazdani
- Department of Medical Biochemistry, Oslo University Hospital, Rikshospitalet, Oslo, 0027, Norway.
| |
Collapse
|
44
|
He F, Yu J, Ma S, Zhao W, Wang Q, He H, Zhang M, Wang J, Lu Z. MiR-34a promotes mitochondrial pathway of apoptosis in human salivary gland epithelial cells by activating NF-κB signaling. Arch Biochem Biophys 2024; 758:110063. [PMID: 38880321 DOI: 10.1016/j.abb.2024.110063] [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: 02/19/2024] [Revised: 05/10/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
To investigate the potential molecular mechanism of miR-34a in Sjögren's syndrome (SS). Transmission electron microscopy was used to observe the salivary gland tissues of mild and severe SS patients. SS mouse model was constructed and injected with miR-34a antagonist. HSGE cells were transfected with miR-34a mimic. Starbase predicted miR-34a binding sites and validated them with dual-luciferase reporter assays. Immunohistochemistry, HE staining, CCK-8, TUNEL assay, flow cytometry, immunofluorescence and Western Blot were used to investigate the effects of miR-34a on NF-κB signaling and mitochondrial pathway of apoptosis in HSGE cells. Severe SS patients showed obvious mitochondrial damage and apoptosis in salivary glands. MiR-34a was overexpressed and NF-κB signaling is activated in salivary glands of severe SS patients. Inhibition of miR-34a alleviated salivary gland injury in SS mice, as well as inhibited the activation of NF-κB signaling and mitochondrial pathway of apoptosis. In conclusion, miR-34a promoted NF-κB signaling by targeting IκBα, thereby causing mitochondrial pathway apoptosis and aggravating SS-induced salivary gland damage.
Collapse
Affiliation(s)
- Fang He
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, China
| | - Juan Yu
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, China
| | - Sha Ma
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, China
| | - Weiqing Zhao
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, China
| | - Qi Wang
- Department of Hematology, The First People's Hospital of Yunnan Province, China
| | - Haitao He
- Department of Hematology, The First People's Hospital of Yunnan Province, China
| | - Mingxing Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, China
| | - Juan Wang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, China
| | - Zhixiang Lu
- Department of Hematology, The First People's Hospital of Yunnan Province, China.
| |
Collapse
|
45
|
Riccio A, Brannon A, Krahn J, Bouvette J, Williams J, Borgnia M, Copeland W. Coordinated DNA polymerization by Polγ and the region of LonP1 regulated proteolysis. Nucleic Acids Res 2024; 52:7863-7875. [PMID: 38932681 PMCID: PMC11260448 DOI: 10.1093/nar/gkae539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
The replicative mitochondrial DNA polymerase, Polγ, and its protein regulation are essential for the integrity of the mitochondrial genome. The intricacies of Polγ regulation and its interactions with regulatory proteins, which are essential for fine-tuning polymerase function, remain poorly understood. Misregulation of the Polγ heterotrimer, consisting of (i) PolG, the polymerase catalytic subunit and (ii) PolG2, the accessory subunit, ultimately results in mitochondrial diseases. Here, we used single particle cryo-electron microscopy to resolve the structure of PolG in its apoprotein state and we captured Polγ at three intermediates within the catalytic cycle: DNA bound, engaged, and an active polymerization state. Chemical crosslinking mass spectrometry, and site-directed mutagenesis uncovered the region of LonP1 engagement of PolG, which promoted proteolysis and regulation of PolG protein levels. PolG2 clinical variants, which disrupted a stable Polγ complex, led to enhanced LonP1-mediated PolG degradation. Overall, this insight into Polγ aids in an understanding of mitochondrial DNA replication and characterizes how machinery of the replication fork may be targeted for proteolytic degradation when improperly functioning.
Collapse
Affiliation(s)
- Amanda A Riccio
- Mitochondrial DNA Replication group, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Asia J Brannon
- Mitochondrial DNA Replication group, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Juno M Krahn
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Jonathan Bouvette
- Molecular Microscopy Consortium, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Jason G Williams
- Mass Spectrometry Research and Support Group, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Mario J Borgnia
- Molecular Microscopy Consortium, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - William C Copeland
- Mitochondrial DNA Replication group, Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| |
Collapse
|
46
|
Xie X, Li J, Zhang X, Mo S, Li A, Sun TY, Xie FY, Luo SM, Wang G, Ou XH, Sun QY, Zhou Q. Endonuclease G is dispensable for sperm mitochondrial DNA elimination during spermatogenesis in mice. Biol Open 2024; 13:bio061730. [PMID: 39373150 PMCID: PMC11554256 DOI: 10.1242/bio.061730] [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: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Maternal inheritance of mitochondrial DNA (mtDNA) is a widespread phenomenon in eukaryotes. Our earlier research indicated that sperm mtDNA is removed prior to fertilization in mice, and Endonuclease G (ENDOG) orchestrates the degradation of sperm mitochondria in Caenorhabditis elegans. However, the mechanisms underlying sperm mtDNA disposal in mammals remain poorly understood. To investigate the potential role of ENDOG in sperm mtDNA elimination, we created Endog knockout (Endog-/-) mice. Our findings revealed that Endog-/- mice maintained normal spermatogenesis and fertility. Most strikingly, we detected no substantial discrepancy in sperm mtDNA copy number between Endog-/- and control mice. Furthermore, we noted that sperm mtDNA copy numbers were unchanged in both less motile and motile sperm isolated by Percoll gradient centrifugation from Endog-/- and control mice. Taken together, our results indicate that ENDOG is not essential for spermatogenesis or the elimination of sperm mtDNA in mice.
Collapse
Affiliation(s)
- Xuefeng Xie
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Jianshuang Li
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou, 510220, Guangdong, China
- The College of Life Science and Technology,Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xue Zhang
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Shaomei Mo
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Ang Li
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Tian-Yi Sun
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Feng-Yun Xie
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Shi-Ming Luo
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Guang Wang
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xiang-Hong Ou
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Qing-Yuan Sun
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Qinghua Zhou
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou, 510220, Guangdong, China
- The College of Life Science and Technology,Jinan University, Guangzhou, 510632, Guangdong, China
| |
Collapse
|
47
|
Bell G, Thoma A, Hargreaves IP, Lightfoot AP. The Role of Mitochondria in Statin-Induced Myopathy. Drug Saf 2024; 47:643-653. [PMID: 38492173 DOI: 10.1007/s40264-024-01413-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
Statins represent the primary therapy for combatting hypercholesterolemia and reducing mortality from cardiovascular events. Despite their pleiotropic effects in lowering cholesterol synthesis, circulating cholesterol, as well as reducing the risk of other systemic diseases, statins have adverse events in a small, but significant, population of treated patients. The most prominent of these adverse effects is statin-induced myopathy, which lacks precise definition but is characterised by elevations in the muscle enzyme creatine kinase alongside musculoskeletal complaints, including pain, weakness and fatigue. The exact aetiology of statin-induced myopathy remains to be elucidated, although impaired mitochondrial function is thought to be an important underlying cause. This may result from or be the consequence of several factors including statin-induced inhibition of coenzyme Q10 (CoQ10) biosynthesis, impaired Ca2+ signalling and modified reactive oxygen species (ROS) generation. The purpose of this review article is to provide an update on the information available linking statin therapy with mitochondrial dysfunction and to outline any mechanistic insights, which may be beneficial in the future treatment of myopathic adverse events.
Collapse
Affiliation(s)
- Gavin Bell
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Anastasia Thoma
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Iain P Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK.
| | - Adam P Lightfoot
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK.
| |
Collapse
|
48
|
Toshima T, Yagi M, Do Y, Hirai H, Kunisaki Y, Kang D, Uchiumi T. Mitochondrial translation failure represses cholesterol gene expression via Pyk2-Gsk3β-Srebp2 axis. Life Sci Alliance 2024; 7:e202302423. [PMID: 38719751 PMCID: PMC11079605 DOI: 10.26508/lsa.202302423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Neurodegenerative diseases and other age-related disorders are closely associated with mitochondrial dysfunction. We previously showed that mice with neuron-specific deficiency of mitochondrial translation exhibit leukoencephalopathy because of demyelination. Reduced cholesterol metabolism has been associated with demyelinating diseases of the brain such as Alzheimer's disease. However, the molecular mechanisms involved and relevance to the pathogenesis remained unknown. In this study, we show that inhibition of mitochondrial translation significantly reduced expression of the cholesterol synthase genes and degraded their sterol-regulated transcription factor, sterol regulatory element-binding protein 2 (Srebp2). Furthermore, the phosphorylation of Pyk2 and Gsk3β was increased in the white matter of p32cKO mice. We observed that Pyk2 inhibitors reduced the phosphorylation of Gsk3β and that GSK3β inhibitors suppressed degradation of the transcription factor Srebp2. The Pyk2-Gsk3β axis is involved in the ubiquitination of Srebp2 and reduced expression of cholesterol gene. These results suggest that inhibition of mitochondrial translation may be a causative mechanism of neurodegenerative diseases of aging. Improving the mitochondrial translation or effectiveness of Gsk3β inhibitors is a potential therapeutic strategy for leukoencephalopathy.
Collapse
Affiliation(s)
- Takahiro Toshima
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yura Do
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
| | - Haruka Hirai
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuya Kunisaki
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
- Kashiigaoka Rehabilitation Hospital, Fukuoka, Japan
- Department of Medical Laboratory Science, Faculty of Health Sciences, Junshin Gakuen University, Fukuoka, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Kyushu University, Fukuoka, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
49
|
Yuan T, Kumar S, Skinner ME, Victor-Joseph R, Abuaita M, Keijer J, Zhang J, Kunkel TJ, Liu Y, Petrunak EM, Saunders TL, Lieberman AP, Stuckey JA, Neamati N, Al-Murshedi F, Alfadhel M, Spelbrink JN, Rodenburg R, de Boer VC, Lombard DB. Human SIRT5 variants with reduced stability and activity do not cause neuropathology in mice. iScience 2024; 27:109991. [PMID: 38846003 PMCID: PMC11154205 DOI: 10.1016/j.isci.2024.109991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/06/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
SIRT5 is a sirtuin deacylase that removes negatively charged lysine modifications, in the mitochondrial matrix and elsewhere in the cell. In benign cells and mouse models, under basal conditions, the phenotypes of SIRT5 deficiency are quite subtle. Here, we identify two homozygous SIRT5 variants in patients suspected to have mitochondrial disease. Both variants, P114T and L128V, are associated with reduced SIRT5 protein stability and impaired biochemical activity, with no evidence of neomorphic or dominant negative properties. The crystal structure of the P114T enzyme was solved and shows only subtle deviations from wild-type. Via CRISPR-Cas9, we generated a mouse model that recapitulates the human P114T mutation; homozygotes show reduced SIRT5 levels and activity, but no obvious metabolic abnormalities, neuropathology, or other gross phenotypes. We conclude that these human SIRT5 variants most likely represent severe hypomorphs, but are likely not by themselves the primary pathogenic cause of the neuropathology observed in the patients.
Collapse
Affiliation(s)
- Taolin Yuan
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, the Netherlands
| | - Surinder Kumar
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mary E. Skinner
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan Victor-Joseph
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Majd Abuaita
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, the Netherlands
| | - Jessica Zhang
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Thaddeus J. Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yanghan Liu
- Department of Medicinal Chemistry, College of Pharmacy and Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elyse M. Petrunak
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas L. Saunders
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Jeanne A. Stuckey
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy and Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fathiya Al-Murshedi
- Genetic and Developmental Medicine Clinic, Department of Genetics, Sultan Qaboos University Hospital, Sultan Qaboos University, Muscat, Oman
| | - Majid Alfadhel
- Medical Genomic Research Department, King Abdullah International Medical Research Center(KAIMRC), King Saud Bin Abdulaziz University for Health Sciences(KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
- Genetics and Precision Medicine Department (GPM), King Abdullah Specialized Children’s Hospital (KASCH), King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
| | - Johannes N. Spelbrink
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Richard Rodenburg
- Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vincent C.J. de Boer
- Human and Animal Physiology, Wageningen University, De Elst 1, Wageningen, the Netherlands
| | - David B. Lombard
- Department of Pathology & Laboratory Medicine, Miller School of Medicine, and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
- Miami VA Healthcare System, Miami, FL 33125, USA
| |
Collapse
|
50
|
Yang Y, Qiu L. Research Progress on the Pathogenesis, Diagnosis, and Drug Therapy of Alzheimer's Disease. Brain Sci 2024; 14:590. [PMID: 38928590 PMCID: PMC11201671 DOI: 10.3390/brainsci14060590] [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: 05/15/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
As the population ages worldwide, Alzheimer's disease (AD), the most prevalent kind of neurodegenerative disorder among older people, has become a significant factor affecting quality of life, public health, and economies. However, the exact pathogenesis of Alzheimer's remains elusive, and existing highly recognized pathogenesis includes the amyloid cascade hypothesis, Tau neurofibrillary tangles hypothesis, and neuroinflammation hypothesis. The major diagnoses of Alzheimer's disease include neuroimaging positron emission computed tomography, magnetic resonance imaging, and cerebrospinal fluid molecular diagnosis. The therapy of Alzheimer's disease primarily relies on drugs, and the approved drugs on the market include acetylcholinesterase drugs, glutamate receptor antagonists, and amyloid-β monoclonal antibodies. Still, the existing drugs can only alleviate the symptoms of the disease and cannot completely reverse it. This review aims to summarize existing research results on Alzheimer's disease pathogenesis, diagnosis, and drug therapy, with the objective of facilitating future research in this area.
Collapse
Affiliation(s)
- Yixuan Yang
- College of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Lina Qiu
- College of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
- Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|