1
|
Qu X, Lai X, He M, Zhang J, Xiang B, Liu C, Huang R, Shi Y, Qiao J. Investigation of epilepsy-related genes in a Drosophila model. Neural Regen Res 2026; 21:195-211. [PMID: 39688550 PMCID: PMC12094548 DOI: 10.4103/nrr.nrr-d-24-00877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/15/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Complex genetic architecture is the major cause of heterogeneity in epilepsy, which poses challenges for accurate diagnosis and precise treatment. A large number of epilepsy candidate genes have been identified from clinical studies, particularly with the widespread use of next-generation sequencing. Validating these candidate genes is emerging as a valuable yet challenging task. Drosophila serves as an ideal animal model for validating candidate genes associated with neurogenetic disorders such as epilepsy, due to its rapid reproduction rate, powerful genetic tools, and efficient use of ethological and electrophysiological assays. Here, we systematically summarize the advantageous techniques of the Drosophila model used to investigate epilepsy genes, including genetic tools for manipulating target gene expression, ethological assays for seizure-like behaviors, electrophysiological techniques, and functional imaging for recording neural activity. We then introduce several typical strategies for identifying epilepsy genes and provide new insights into gene‒gene interactions in epilepsy with polygenic causes. We summarize well-established precision medicine strategies for epilepsy and discuss prospective treatment options, including drug therapy and gene therapy for genetic epilepsy based on the Drosophila model. Finally, we also address genetic counseling and assisted reproductive technology as potential approaches for the prevention of genetic epilepsy.
Collapse
Affiliation(s)
- Xiaochong Qu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xiaodan Lai
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Mingfeng He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Jinyuan Zhang
- School of Health Management, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Binbin Xiang
- The First Clinical Medicine School of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Chuqiao Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Ruina Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yiwu Shi
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Jingda Qiao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Liu H, Shi W, Zhang X, He X, Zhao X. Identifying sex-based disparities in porcine mitochondrial function. Anim Biotechnol 2025; 36:2488068. [PMID: 40208306 DOI: 10.1080/10495398.2025.2488068] [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: 09/01/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
In pigs, the effect of sex on production and reproductive traits has been largely reported, however, whether sex exerts its influence through regulating mitochondrial function is still unclear. In this study, we constructed 15 male cells and 15 female fibroblasts derived from 35-day and 50-day fetuses, newborn piglets and 1-year-old pigs to identify the sex effect on mitochondrial functions. Results indicated significant differences on cellular and molecular characteristics between male and female cells, including energy metabolic trait, mitochondrial DNA (mtDNA) replication and transcription, and mRNA expressions of mitochondrial biogenesis genes and mitoprotease genes. Referring to sex, males exhibited significantly higher oxygen consumption rate productions, levels of reactive oxygen species (ROS) and mtDNA copy numbers than those with females in muscle and ear fibroblasts. And the expressions of mtDNA, mitochondrial biogenesis genes (POLG, PPARGC1A, TFAM and TWNK) and XPNPEP3 were higher in males than females in ear fibroblasts derived from 1-year-old adult pigs (EFA cells). While, the cell proliferation and expressions of genes related to ROS metabolism were not influenced by sex. The results highlight the effect of sex on mitochondrial function and gene expression, and provide important data for a comprehensive understanding of the mechanisms underlying sex regulation of energy metabolism-related traits in pigs.
Collapse
Affiliation(s)
- Hao Liu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenshu Shi
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
| | - Xing Zhang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
| | - Xinmiao He
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, China
| | - Xingbo Zhao
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Parchwani D, Singh R, Patel D. Biological and translational attributes of mitochondrial DNA copy number: Laboratory perspective to clinical relevance. World J Methodol 2025; 15:102709. [DOI: 10.5662/wjm.v15.i3.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/21/2025] [Accepted: 02/08/2025] [Indexed: 03/06/2025] Open
Abstract
The mitochondrial DNA copy number (mtDNAcn) plays a vital role in cellular energy metabolism and mitochondrial health. As mitochondria are responsible for adenosine triphosphate production through oxidative phosphorylation, maintaining an appropriate mtDNAcn level is vital for the overall cellular function. Alterations in mtDNAcn have been linked to various diseases, including neurodegenerative disorders, metabolic conditions, and cancers, making it an important biomarker for understanding the disease pathogenesis. The accurate estimation of mtDNAcn is essential for clinical applications. Quantitative polymerase chain reaction and next-generation sequencing are commonly employed techniques with distinct advantages and limitations. Clinically, mtDNAcn serves as a valuable indicator for early diagnosis, disease progression, and treatment response. For instance, in oncology, elevated mtDNAcn levels in blood samples are associated with tumor aggressiveness and can aid in monitoring treatment efficacy. In neurodegenerative diseases such as Alzheimer’s and Parkinson’s, altered mtDNAcn patterns provide insights into disease mechanisms and progression. Understanding and estimating mtDNAcn are critical for advancing diagnostic and therapeutic strategies in various medical fields. As research continues to uncover the implications of mtDNAcn alterations, its potential as a clinical biomarker is likely to expand, thereby enhancing our ability to diagnose and manage complex diseases.
Collapse
Affiliation(s)
- Deepak Parchwani
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot 360001, India
| | - Ragini Singh
- Department of Biochemistry, All India Institute of Medical Sciences, Rajkot 360001, India
| | - Digisha Patel
- Department of Physiology, Shantabaa Medical College and General Hospital Amreli, Amreli 365601, Gujarāt, India
| |
Collapse
|
4
|
McQuillin A, Ophoff RA. Genomics of Bipolar Disorder: What the Clinician Needs to Know. Psychiatr Clin North Am 2025; 48:331-341. [PMID: 40348421 DOI: 10.1016/j.psc.2025.01.009] [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] [Indexed: 05/14/2025]
Abstract
Bipolar disorder (BD) affects approximately 2% of the global population, characterized by alternating episodes of mania or hypomania, and depression. It comprises two main types: bipolar I disorder, marked by severe manic episodes, and bipolar II disorder, defined by milder hypomanic episodes. Individuals often experience rapid cycling and significant comorbidities, leading to decreased productivity and increased mortality rates. Early diagnosis and intervention are crucial for better outcomes. Both genetic and environmental factors contribute to BD's etiology, with genetic research promising improved diagnosis, novel therapeutic targets, and societal understanding that may help destigmatize the disorder.
Collapse
Affiliation(s)
- Andrew McQuillin
- Neuroscience Mental Health Department, Division of Psychiatry, University College London, Gower Street, London, WC1E 6BT, UK
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Moe A, Fry C, Baker MR, Ng YS. A 38-year-old man with unsteadiness and difficulty walking. Pract Neurol 2025:pn-2024-004352. [PMID: 40409782 DOI: 10.1136/pn-2024-004352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2025] [Indexed: 05/25/2025]
Affiliation(s)
- Aye Moe
- Directorate of Neurosciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Newcastle University, Newcastle upon Tyne, UK
| | - Charles Fry
- Department of Clinical Neurophysiology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Mark R Baker
- Directorate of Neurosciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Newcastle University, Newcastle upon Tyne, UK
- Department of Clinical Neurophysiology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Yi Shiau Ng
- Directorate of Neurosciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
6
|
Corrà S, Zuppardo A, Valenzuela S, Jenninger L, Cerutti R, Sillamaa S, Hoberg E, Johansson KAS, Rovsnik U, Volta S, Silva-Pinheiro P, Davis H, Trifunovic A, Minczuk M, Gustafsson CM, Suomalainen A, Zeviani M, Macao B, Zhu X, Falkenberg M, Viscomi C. Modelling POLG mutations in mice unravels a critical role of POLγΒ in regulating phenotypic severity. Nat Commun 2025; 16:4782. [PMID: 40404629 PMCID: PMC12098916 DOI: 10.1038/s41467-025-60059-y] [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: 08/26/2024] [Accepted: 05/14/2025] [Indexed: 05/24/2025] Open
Abstract
DNA polymerase γ (POLγ), responsible for mitochondrial DNA replication, consists of a catalytic POLγA subunit and two accessory POLγB subunits. Mutations in POLG, which encodes POLγA, lead to various mitochondrial diseases. We investigated the most common POLG mutations (A467T, W748S, G848S, Y955C) by characterizing human and mouse POLγ variants. Our data reveal that these mutations significantly impair POLγ activities, with mouse variants exhibiting milder defects. Cryogenic electron microscopy highlighted structural differences between human and mouse POLγ, particularly in the POLγB subunit, which may explain the higher activity of mouse POLγ and the reduced severity of mutations in mice. We further generated a panel of mouse models mirroring common human POLG mutations, providing crucial insights into the pathogenesis of POLG-related disorders and establishing robust models for therapeutic development. Our findings emphasize the importance of POLγB in modulating the severity of POLG mutations.
Collapse
Affiliation(s)
- Samantha Corrà
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
| | - Alessandro Zuppardo
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy
| | - Sebastian Valenzuela
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Louise Jenninger
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Raffaele Cerutti
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy
| | - Sirelin Sillamaa
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Emily Hoberg
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Katarina A S Johansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Urska Rovsnik
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Sara Volta
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy
| | - Pedro Silva-Pinheiro
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Hannah Davis
- The Mary Lyon Centre, MRC Harwell, Becquerel Ave, Didcot, Oxfordshire, OX11 0RD, UK
| | - Aleksandra Trifunovic
- Institute for Mitochondrial Diseases and Aging, Faculty of Medicine, CECAD Research Center, 50931, Cologne, Germany
| | - Michal Minczuk
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 2PY, UK
| | - Claes M Gustafsson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Anu Suomalainen
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, Haartmaninkatu 8, University of Helsinki, 00290, Helsinki, Finland
- HUSlab, Helsinki University Hospital, University of Helsinki, 00290, Helsinki, Finland
| | - Massimo Zeviani
- Department of Neurosciences, University of Padova, Via Belzoni 160, 35121, Padova, Italy
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Via Istria 61, 34137, Trieste, Italy
| | - Bertil Macao
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden
| | - Xuefeng Zhu
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden.
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China.
- National Key Laboratory for Development and Utilization of Forest Food Resources, Zhejiang A&F University, Hangzhou, 311300, China.
| | - Maria Falkenberg
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Medicinaregatan 9A, P.O. Box 440, 41390, Gothenburg, Sweden.
| | - Carlo Viscomi
- Veneto Institute of Molecular Medicine (VIMM), Via Orus, 2-35129, Padova, Italy.
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B-35131, Padova, Italy.
| |
Collapse
|
7
|
Yang Y, Li X, Xiao S, Wei Q, Ren L, Yao Y, Liu N. PARylation of POLG Mediated by PARP1 Accelerates Ferroptosis-Induced Vascular Calcification via Activating Adora2a/Rap1 Signaling. Arterioscler Thromb Vasc Biol 2025. [PMID: 40401372 DOI: 10.1161/atvbaha.124.321682] [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: 12/31/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Vascular calcification (VC) is associated with diabetes, chronic kidney disease, and aging. VC is found to be a powerful and independent risk factor for cardiovascular mortality. Vascular smooth muscle cell (VSMC) ferroptosis, a form of cell death, is known to be involved in VC. However, whether VSMC ferroptosis is regulated by posttranslational modifications remains undefined. METHODS We explored the potential role and mechanism of PARP1 (poly[ADP-ribose] polymerase 1)-mediated poly(ADP-ribosyl)ation (PARylation) in VSMC ferroptosis during VC. Mouse VSMCs were treated with β-glycerophosphate, and Parp1flox/flox Tagln Cre+ calcified mice were generated with AAV9-sh-POLG (DNA polymerase gamma) injected to establish in vitro and in vivo models, respectively. RNA-sequencing analysis was performed to determine the transcriptomic alterations in VSMCs overexpressing POLG and treated with β-glycerophosphate. RESULTS Both PARP1 expression and PARylation levels were increased in β-glycerophosphate-induced VC, with PARP1 knockdown mitigating VC by improving mitochondrial function and inhibiting the subsequent VSMC ferroptosis. Mechanistically, POLG PARylation levels were increased in calcified VSMCs from PARP1 activation, triggering PARylation-dependent ubiquitination of POLG that resulted in POLG downregulation. This led to mitochondrial dysfunction and Adora2a (adenosine receptor A2A)/Rap1 (Ras-associated protein 1) signaling pathway activation to induce VSMC ferroptosis, which ultimately aggravated VC. CONCLUSIONS Our study establishes the critical role of PARP1-mediated PARylation-dependent ubiquitination of POLG in VSMC ferroptosis-induced VC. These findings suggest that PARP1 inhibitors could potentially serve as novel therapeutic strategies for VC.
Collapse
Affiliation(s)
- Yiqing Yang
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xiaoxue Li
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Shengjue Xiao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Qin Wei
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Liqun Ren
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Naifeng Liu
- Department of Cardiology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
8
|
VanPortfliet JJ, Lei Y, Ramanathan M, Martinez CG, Wong J, Stodola TJ, Hoffmann BR, Pflug K, Sitcheran R, Kneeland SC, Murray SA, McGuire PJ, Cannon CL, West AP. Caspase-11 drives macrophage hyperinflammation in models of Polg-related mitochondrial disease. Nat Commun 2025; 16:4640. [PMID: 40393978 PMCID: PMC12092707 DOI: 10.1038/s41467-025-59907-8] [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: 05/13/2024] [Accepted: 05/06/2025] [Indexed: 05/22/2025] Open
Abstract
Mitochondrial diseases (MtD) represent a significant public health challenge due to their heterogenous clinical presentation, often severe and progressive symptoms, and lack of effective therapies. Environmental exposures, such bacterial and viral infection, can further compromise mitochondrial function and exacerbate the progression of MtD. However, the underlying immune alterations that enhance immunopathology in MtD remain unclear. Here we employ in vitro and in vivo approaches to clarify the molecular and cellular basis for innate immune hyperactivity in models of polymerase gamma (Polg)-related MtD. We reveal that type I interferon (IFN-I)-mediated upregulation of caspase-11 and guanylate-binding proteins (GBP) increase macrophage sensing of the opportunistic microbe Pseudomonas aeruginosa (PA) in Polg mutant mice. Furthermore, we show that excessive cytokine secretion and activation of pyroptotic cell death pathways contribute to lung inflammation and morbidity after infection with PA. Our work provides a mechanistic framework for understanding innate immune dysregulation in MtD and reveals potential targets for limiting infection- and inflammation-related complications in Polg-related MtD.
Collapse
Affiliation(s)
- Jordyn J VanPortfliet
- The Jackson Laboratory, Bar Harbor, ME, USA
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Yuanjiu Lei
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | | | - Camila Guerra Martinez
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA
| | | | | | | | - Kathryn Pflug
- Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Raquel Sitcheran
- Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | | | | | - Peter J McGuire
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carolyn L Cannon
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - A Phillip West
- The Jackson Laboratory, Bar Harbor, ME, USA.
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|
9
|
Smith LA, Keane EB, Connor K, Chan F, Cunningham MO. In vitro modelling of the neuropathophysiological features of mitochondrial epilepsy. Seizure 2025:S1059-1311(25)00121-9. [PMID: 40410091 DOI: 10.1016/j.seizure.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/25/2025] Open
Abstract
Epilepsy is a common and severe neurological manifestation of primary mitochondrial disease, affecting approximately 60 % of paediatric patients and 20 % of adult patients. Many of the mitochondrial epilepsies, particularly those presenting in childhood, are refractory to anti-epileptic treatment. Moreover, these conditions are typically characterised by severe neurodegeneration and closely associated with neurological decline and premature death. Indeed, there persists an urgent need to delineate the mechanisms underpinning mitochondrial epilepsy in order to develop effective treatments. In this review, we provide an overview of currently available in vitro models of the mitochondrial epilepsies. Such models offer opportunities to characterise early disease pathophysiology and interrogate novel mitochondrial-targeting and anti-epileptic treatments, with an overall aim to modulate seizure associated pathology and activity for the mitochondrial epilepsies. We discuss the use of acute cortical and subcortical brain slice preparations, obtained from both neurosurgical patients and rodents, for modelling the common neuropathophysiological features of mitochondrial epilepsy. We also review the use of induced pluripotent stem cell derived neural and glial culture models, and the development of three-dimensional cerebral organoids, generated from fibroblasts obtained from patients with primary mitochondrial disease. Human-derived, disease-relevant in vitro model systems which recapitulate the complexity and pathological features observed in patient brain tissues are crucial to help bridge the gap between animal models and patients living with mitochondrial epilepsy.
Collapse
Affiliation(s)
- Laura A Smith
- Mitochondrial Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - Ella B Keane
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Ireland
| | - Kate Connor
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Ireland
| | - Felix Chan
- Department of Pharmacy, School of Health Sciences, University of Birmingham, Birmingham, B15 2TT, UK; Centre for Human Brain Health (CHBH), University of Birmingham, Birmingham, B15 2TT, UK; Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, B15 2TT, UK
| | - Mark O Cunningham
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Ireland.
| |
Collapse
|
10
|
Varughese R, Rahman S. Endocrine Dysfunction in Primary Mitochondrial Diseases. Endocr Rev 2025; 46:376-396. [PMID: 39891580 PMCID: PMC12063101 DOI: 10.1210/endrev/bnaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/30/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Primary mitochondrial disorders (PMD) are genetic disorders affecting the structure or function of the mitochondrion. Mitochondrial functions are diverse, including energy production, ion homeostasis, reactive oxygen species regulation, antioxidant defense, and biosynthetic responsibilities, notably including steroidogenesis. Mitochondria provide the energy to drive intracellular production and extracellular secretion of all hormones. The understanding of the endocrine consequences of PMD is key to timely identification of both endocrine complications in PMD patients, and PMD presenting primarily with endocrine disease. This is a narrative review on the endocrine manifestations of PMD, underlying disease mechanisms, and current and emerging approaches to diagnosing and treating these complex disorders. Diabetes is the most frequent endocrine manifestation of PMD, but growth hormone deficiency, adrenal insufficiency, hypogonadism, and parathyroid dysfunction may occur. Despite the intricate involvement of the thyroid gland in metabolic regulation, there is little evidence for a causal relationship between thyroid dysfunction and PMD. In conclusion, endocrine dysfunction is observed in PMD with varying incidence depending on the specific mitochondrial disorder and the endocrine organ in question. Diagnosis of PMD in a patient with endocrine-presenting features requires a high level of clinical suspicion, particularly when apparently unrelated comorbidities co-exist. Similarly, endocrine pathology may be subtle in patients with known PMD, and thorough consideration must be given to ensure timely diagnosis and treatment. The scope for novel therapeutics for this group of devastating conditions is enormous; however, several challenges remain to be overcome before hopes of curative treatments can be brought into clinical practice.
Collapse
Affiliation(s)
- Rachel Varughese
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Shamima Rahman
- Mitochondrial Research Group, Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Metabolic Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| |
Collapse
|
11
|
Shan M, Xu L, Yang W, Sui L, Sun P, Zhuo X, Liu S. Identification of common hub genes and construction of immune regulatory networks in aplastic anemia, myelodysplastic syndromes, and acute myeloid leukemia. Front Immunol 2025; 16:1547289. [PMID: 40406144 PMCID: PMC12095185 DOI: 10.3389/fimmu.2025.1547289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Background Aplastic anemia (AA), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML) exhibit complex pathogenic mechanisms and interrelated characteristics. We aimed to identify the common hub genes, establishing a foundation for preventing disease progression. Methods We selected relevant datasets from the Gene Expression Omnibus(GEO) database for differential gene expression, gene set enrichment, and weighted gene co-expression network analyses to identify hub genes, and then validated them. Subsequent analyses included immune infiltration analysis, single-cell sequencing, and cell communication analysis. We performed Mendelian randomization to screen inflammatory factors and immune cells. We used RT-qPCR, Enzyme - Linked Immunosorbent Assay(ELISA), and cell proliferation assays to validate the identified hub genes, their relationship with cellular communication mediators and inflammatory factors, and their impact on cellular function. Results POLG and MAP2K7 were identified as common hub genes, with low expression observed across AA, MDS, and AML. There were distinct immune differentials among these diseases, with an enhanced correlation between immune cells and hub genes as the disease progressed. Macrophage Migration Inhibitory Factor(MIF) emerged as a key mediator of cellular communication. We identified 20 regulatory pathways of immune cells and inflammatory factors across different disease stages. In vitro validation confirmed low expression of the hub genes, which were inversely correlated with MIF and inflammatory factors, though they showed no significant impact on cell proliferation or migration. Conclusions POLG and MAP2K7 demonstrate crucial roles in the progression from AA to MDS and, ultimately, to AML. These genes regulate more than 20 immune regulatory pathways through MIF-mediated communication, thereby influencing disease progression.
Collapse
Affiliation(s)
- Mingliang Shan
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Li Xu
- School of Management, Shandong Second Medical University, Weifang, China
| | - Wenzhe Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lili Sui
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Ping Sun
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Xiumei Zhuo
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Lucchesi M, Biso L, Bonaso M, Longoni B, Buchignani B, Battini R, Santorelli FM, Doccini S, Scarselli M. Mitochondrial Dysfunction in Genetic and Non-Genetic Parkinson's Disease. Int J Mol Sci 2025; 26:4451. [PMID: 40362688 PMCID: PMC12072996 DOI: 10.3390/ijms26094451] [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: 03/21/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondrial dysfunction is a hallmark of Parkinson's disease (PD) pathogenesis, contributing to increased oxidative stress and impaired endo-lysosomal-proteasome system efficiency underlying neuronal injury. Genetic studies have identified 19 monogenic mutations-accounting for ~10% of PD cases-that affect mitochondrial function and are associated with early- or late-onset PD. Early-onset forms typically involve genes encoding proteins essential for mitochondrial quality control, including mitophagy and structural maintenance, while late-onset mutations impair mitochondrial dynamics, bioenergetics, and trafficking. Atypical juvenile genetic syndromes also exhibit mitochondrial abnormalities. In idiopathic PD, environmental neurotoxins such as pesticides and MPTP act as mitochondrial inhibitors, disrupting complex I activity and increasing reactive oxygen species. These converging pathways underscore mitochondria as a central node in PD pathology. This review explores the overlapping and distinct mitochondrial mechanisms in genetic and non-genetic PD, emphasizing their role in neuronal vulnerability. Targeting mitochondrial dysfunction finally offers a promising therapeutic avenue to slow or modify disease progression by intervening at a key point of neurodegenerative convergence.
Collapse
Affiliation(s)
| | - Letizia Biso
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| | - Marco Bonaso
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| | - Biancamaria Longoni
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| | - Bianca Buchignani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Stefano Doccini
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Marco Scarselli
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.B.); (B.L.); (B.B.); (M.S.)
| |
Collapse
|
13
|
Kielty K, Collyer J, Ganesh KD, Narayanan S, Rajan DS. Spectrum of clinical neuroimaging in mitochondrial disorders: a neuroanatomical approach. Pediatr Radiol 2025:10.1007/s00247-025-06252-z. [PMID: 40316773 DOI: 10.1007/s00247-025-06252-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 05/04/2025]
Abstract
Mitochondrial disorders are a highly heterogeneous group of genetic diseases that impact pathways associated with the structure and function of the mitochondrion. Clinical presentations of mitochondrial disorders include a wide range of onset, progression, and spectrum of neurological symptoms - ranging from episodic, focal neurological deficits to gradual onset of developmental delays, sensorineural hearing loss, visual impairment, or ataxia. This variability provides clinicians with a diagnostic challenge in identifying suspicion of a mitochondrial disorder and prioritizing specific mitochondrial disorders within their differential. While next-generation sequencing of both the nuclear and mitochondrial genomes has aided identification of mitochondrial disorders, testing results are typically not available for weeks to months, and CSF and biochemical studies indicating possible mitochondrial disorder, such as elevated lactate, are nonspecific in differentiating between mitochondrial disorders and other neurogenetic diseases. Neuroimaging can serve as an early tool to help identify specific mitochondrial disorders; however, there are additional variability and overlap between disorders and other non-mitochondrial diseases. This review provides a framework in narrowing the mitochondrial differential by neuroanatomical localization on neuroimaging studies. We will highlight established neuroimaging patterns associated with mitochondrial disorders, review the role of MRS, and discuss the alternative non-mitochondrial etiologies associated with these findings.
Collapse
Affiliation(s)
- Kate Kielty
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
- University of Pittsburgh, Pittsburgh, PA, USA.
| | - John Collyer
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- University of Pittsburgh, Pittsburgh, PA, USA
| | - Krrithvi Dharini Ganesh
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- University of Pittsburgh, Pittsburgh, PA, USA
| | - Srikala Narayanan
- Texas Children's Hospital, Houston, TX, USA
- Baylor College of Medicine, Houston, TX, USA
| | - Deepa S Rajan
- Division of Child Neurology, Department of Pediatrics, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Peacock DJSJ, Ferreira CR, Horvath G, Hoffmann GF, Blau N, Ebrahimi-Fakhari D. Clinical and biochemical footprints of inherited metabolic diseases: Ia. Movement disorders, updated. Mol Genet Metab 2025; 145:109084. [PMID: 40132382 DOI: 10.1016/j.ymgme.2025.109084] [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: 12/20/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Movement disorders are a common manifestation of inherited metabolic diseases (IMDs), categorized into hyperkinetic movement disorders, hypokinetic-rigid syndromes, ataxia, and spasticity. We reviewed and updated the list of known metabolic disorders associated with movement disorders, identifying a total of 559 IMDs. We outlined the more common and treatable causes, sorted by the dominant movement disorder phenomenology, and provided a practical clinical approach for suspected IMDs presenting with movement disorders. This work represents an updated catalog in a series of articles aimed at creating and maintaining a comprehensive list of clinical and metabolic differential diagnoses based on system involvement.
Collapse
Affiliation(s)
- Dakota J S J Peacock
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, Canada
| | - Carlos R Ferreira
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| | - Gabriella Horvath
- Division of Biochemical Genetics, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, Canada
| | - Georg F Hoffmann
- University Children's Hospital, University Clinic Heidelberg, Germany
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital, Zürich, Switzerland.
| | - Darius Ebrahimi-Fakhari
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Carvalho G, Nguyen TVH, Repolês B, Forslund JME, Wijethunga WMRR, Ranjbarian F, Mendes IC, Gorospe CM, Chaudhari N, Falabella M, Doimo M, Wanrooij S, Pitceathly RDS, Hofer A, Wanrooij PH. Activating AMPK improves pathological phenotypes due to mtDNA depletion. FEBS J 2025; 292:2359-2380. [PMID: 39918244 PMCID: PMC12062783 DOI: 10.1111/febs.70006] [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: 07/02/2024] [Revised: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 05/11/2025]
Abstract
AMP-activated protein kinase (AMPK) is a master regulator of cellular energy homeostasis that also plays a role in preserving mitochondrial function and integrity. Upon a disturbance in the cellular energy state that increases AMP levels, AMPK activity promotes a switch from anabolic to catabolic metabolism to restore energy homeostasis. However, the level of severity of mitochondrial dysfunction required to trigger AMPK activation is currently unclear, as is whether stimulation of AMPK using specific agonists can improve the cellular phenotype following mitochondrial dysfunction. Using a cellular model of mitochondrial disease characterized by progressive mitochondrial DNA (mtDNA) depletion and deteriorating mitochondrial metabolism, we show that mitochondria-associated AMPK becomes activated early in the course of the advancing mitochondrial dysfunction, before any quantifiable decrease in the ATP/(AMP + ADP) ratio or respiratory chain activity. Moreover, stimulation of AMPK activity using the specific small-molecule agonist A-769662 alleviated the mitochondrial phenotypes caused by the mtDNA depletion and restored normal mitochondrial membrane potential. Notably, the agonist treatment was able to partially restore mtDNA levels in cells with severe mtDNA depletion, while it had no impact on mtDNA levels of control cells. The beneficial impact of the agonist on mitochondrial membrane potential was also observed in cells from patients suffering from mtDNA depletion. These findings improve our understanding of the effects of specific small-molecule activators of AMPK on mitochondrial and cellular function and suggest a potential application for these compounds in disease states involving mtDNA depletion.
Collapse
Affiliation(s)
- Gustavo Carvalho
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Tran V. H. Nguyen
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Bruno Repolês
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | | | | | | | - Isabela C. Mendes
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | | | - Namrata Chaudhari
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Micol Falabella
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUK
| | - Mara Doimo
- Clinical Genetics Unit, Department of Women and Children's HealthPadua UniversityPaduaItaly
| | - Sjoerd Wanrooij
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | - Robert D. S. Pitceathly
- Department of Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular DiseasesThe National Hospital for Neurology and NeurosurgeryLondonUK
| | - Anders Hofer
- Department of Medical Biochemistry and BiophysicsUmeå UniversityUmeåSweden
| | | |
Collapse
|
16
|
Forslund JME, Nguyen TVH, Parkash V, Berner A, Goffart S, Pohjoismäki JLO, Wanrooij PH, Johansson E, Wanrooij S. The POLγ Y951N patient mutation disrupts the switch between DNA synthesis and proofreading, triggering mitochondrial DNA instability. Proc Natl Acad Sci U S A 2025; 122:e2417477122. [PMID: 40238457 PMCID: PMC12036981 DOI: 10.1073/pnas.2417477122] [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: 08/30/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Mitochondrial DNA (mtDNA) stability, essential for cellular energy production, relies on DNA polymerase gamma (POLγ). Here, we show that the POLγ Y951N disease-causing mutation induces replication stalling and severe mtDNA depletion. However, unlike other POLγ disease-causing mutations, Y951N does not directly impair exonuclease activity and only mildly affects polymerase activity. Instead, we found that Y951N compromises the enzyme's ability to efficiently toggle between DNA synthesis and degradation, and is thus a patient-derived mutation with impaired polymerase-exonuclease switching. These findings provide insights into the intramolecular switch when POLγ proofreads the newly synthesized DNA strand and reveal a new mechanism for causing mitochondrial DNA instability.
Collapse
Affiliation(s)
| | - Tran V. H. Nguyen
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå90187, Sweden
| | - Vimal Parkash
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå90187, Sweden
| | - Andreas Berner
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå90187, Sweden
| | - Steffi Goffart
- Department of Environmental and Biological Sciences, University of Eastern Finland, JoensuuFI-80101, Finland
| | - Jaakko L. O. Pohjoismäki
- Department of Environmental and Biological Sciences, University of Eastern Finland, JoensuuFI-80101, Finland
| | - Paulina H. Wanrooij
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå90187, Sweden
| | - Erik Johansson
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå90187, Sweden
| | - Sjoerd Wanrooij
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå90187, Sweden
| |
Collapse
|
17
|
Giordano L, Ware SA, Lagranha CJ, Kaufman BA. Mitochondrial DNA signals driving immune responses: Why, How, Where? Cell Commun Signal 2025; 23:192. [PMID: 40264103 PMCID: PMC12012978 DOI: 10.1186/s12964-025-02042-0] [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: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
There has been a recent expansion in our understanding of DNA-sensing mechanisms. Mitochondrial dysfunction, oxidative and proteostatic stresses, instability and impaired disposal of nucleoids cause the release of mitochondrial DNA (mtDNA) from the mitochondria in several human diseases, as well as in cell culture and animal models. Mitochondrial DNA mislocalized to the cytosol and/or the extracellular compartments can trigger innate immune and inflammation responses by binding DNA-sensing receptors (DSRs). Here, we define the features that make mtDNA highly immunogenic and the mechanisms of its release from the mitochondria into the cytosol and the extracellular compartments. We describe the major DSRs that bind mtDNA such as cyclic guanosine-monophosphate-adenosine-monophosphate synthase (cGAS), Z-DNA-binding protein 1 (ZBP1), NOD-, LRR-, and PYD- domain-containing protein 3 receptor (NLRP3), absent in melanoma 2 (AIM2) and toll-like receptor 9 (TLR9), and their downstream signaling cascades. We summarize the key findings, novelties, and gaps of mislocalized mtDNA as a driving signal of immune responses in vascular, metabolic, kidney, lung, and neurodegenerative diseases, as well as viral and bacterial infections. Finally, we define common strategies to induce or inhibit mtDNA release and propose challenges to advance the field.
Collapse
Affiliation(s)
- Luca Giordano
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus-Liebig-University, Giessen, Germany.
| | - Sarah A Ware
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia J Lagranha
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brett A Kaufman
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Valenzuela S, Zhu X, Macao B, Stamgren M, Geukens C, Charifson PS, Kern G, Hoberg E, Jenninger L, Gruszczyk AV, Lee S, Johansson KAS, Miralles Fusté J, Shi Y, Kerns SJ, Arabanian L, Martinez Botella G, Ekström S, Green J, Griffin AM, Pardo-Hernández C, Keating TA, Küppers-Munther B, Larsson NG, Phan C, Posse V, Jones JE, Xie X, Giroux S, Gustafsson CM, Falkenberg M. Small molecules restore mutant mitochondrial DNA polymerase activity. Nature 2025:10.1038/s41586-025-08856-9. [PMID: 40205042 DOI: 10.1038/s41586-025-08856-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/03/2025] [Indexed: 04/11/2025]
Abstract
Mammalian mitochondrial DNA (mtDNA) is replicated by DNA polymerase γ (POLγ), a heterotrimeric complex consisting of a catalytic POLγA subunit and two accessory POLγB subunits1. More than 300 mutations in POLG, the gene encoding the catalytic subunit, have been linked to severe, progressive conditions with high rates of morbidity and mortality, for which no treatment exists2. Here we report on the discovery and characterization of PZL-A, a first-in-class small-molecule activator of mtDNA synthesis that is capable of restoring function to the most common mutant variants of POLγ. PZL-A binds to an allosteric site at the interface between the catalytic POLγA subunit and the proximal POLγB subunit, a region that is unaffected by nearly all disease-causing mutations. The compound restores wild-type-like activity to mutant forms of POLγ in vitro and activates mtDNA synthesis in cells from paediatric patients with lethal POLG disease, thereby enhancing biogenesis of the oxidative phosphorylation machinery and cellular respiration. Our work demonstrates that a small molecule can restore function to mutant DNA polymerases, offering a promising avenue for treating POLG disorders and other severe conditions linked to depletion of mtDNA.
Collapse
Affiliation(s)
- Sebastian Valenzuela
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Xuefeng Zhu
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Bertil Macao
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | - Emily Hoberg
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Louise Jenninger
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Seoeun Lee
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Katarina A S Johansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | - Nils-Göran Larsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Xie Xie
- Pretzel Therapeutics, Mölndal, Sweden
| | | | - Claes M Gustafsson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden.
| | - Maria Falkenberg
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
19
|
Berrahmoune S, Dassi C, Pekeles H, Cheung ACT, Gagnon T, Waters PJ, Buhas D, Myers KA. Investigating the safety and efficacy of deoxycytidine/deoxythymidine in mitochondrial DNA depletion disorders: phase 2 open-label trial. J Neurol 2025; 272:307. [PMID: 40175578 DOI: 10.1007/s00415-025-13060-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
OBJECTIVE Mitochondrial DNA depletion disorders are rare genetic disorders involving mitochondrial dysfunction. These diseases are genetically and clinically heterogeneous but share the common feature of progressively degenerative courses. At present, there are no approved treatments for mitochondrial DNA depletion disorders, though recent reports have suggested that treatment with deoxycytidine/deoxythymidine could be effective for subtypes caused by pathogenic variants in two specific genes, POLG and TK2. We investigated the therapeutic potential of deoxycytidine/deoxythymidine for people with mitochondrial DNA depletion disorders due to pathogenic variants in genes other than POLG and TK2. METHODS We analyzed interim data from an open-label clinical trial of deoxycytidine/deoxythymidine for treatment of mitochondrial DNA depletion disorders, specifically examining disorders due to pathogenic variants in genes other than POLG and TK2. Outcome measures included Newcastle Mitochondrial Disease Scale score and serum growth differentiation factor 15, a mitochondrial function biomarker. RESULTS Data were available from eight individuals having pathogenic variants in FBXL4, SUCLG1, SUCLA2, or RRM2B. Newcastle Mitochondrial Disease Scale score improved in all individuals except for one who withdrew before the first follow-up visit; group level analysis was significant at 1-month and 6-month timepoints. Five patients had elevated growth differentiation factor 15 at baseline; of these, levels improved in four, including three whose values normalized. CONCLUSION These data suggest deoxycytidine/deoxythymidine is a safe and therapeutically promising intervention for a broad range of mitochondrial DNA depletion disorders.
Collapse
Affiliation(s)
- Saoussen Berrahmoune
- Research Institute of the McGill University Health Centre, 2155 Guy Street, Suite 500, Montreal, QC, H3H 2R9, Canada
| | - Christelle Dassi
- Research Institute of the McGill University Health Centre, 2155 Guy Street, Suite 500, Montreal, QC, H3H 2R9, Canada
| | - Heather Pekeles
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, 1001 Décarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Anthony C T Cheung
- Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre, 1001 Décarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Tommy Gagnon
- Medical Genetics Service, Department of Laboratory Medicine, CHUS and Department of Pediatrics, Université de Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada
| | - Paula J Waters
- Medical Genetics Service, Department of Laboratory Medicine, CHUS and Department of Pediatrics, Université de Sherbrooke, Sherbrooke, QC, J1K 2R1, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), 12E Avenue N Porte 6, Sherbrooke, QC, J1H 5N4, Canada
| | - Daniela Buhas
- Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre, 1001 Décarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Kenneth A Myers
- Research Institute of the McGill University Health Centre, 2155 Guy Street, Suite 500, Montreal, QC, H3H 2R9, Canada.
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, 1001 Décarie Boulevard, Montreal, QC, H4A 3J1, Canada.
- Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, 1001 Décarie Boulevard, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
20
|
Kandemirli SG, Al-Dasuqi K, Aslan B, Goldstein A, Alves CAPF. Overview of neuroimaging in primary mitochondrial disorders. Pediatr Radiol 2025; 55:765-791. [PMID: 39937244 DOI: 10.1007/s00247-025-06172-y] [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: 09/06/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 02/13/2025]
Abstract
Advancements in understanding the clinical, biochemical, and genetic aspects of primary mitochondrial disorders, along with the identification of a broad range of phenotypes frequently involving the central nervous system, have opened a new and crucial area in neuroimaging. This expanding knowledge presents significant challenges for radiologists in clinical settings, as the neuroimaging features and their associated metabolic abnormalities become more complex. This review offers a comprehensive overview of the key neuroimaging features associated with the common primary mitochondrial disorders. It highlights both the classical imaging findings and the emerging diagnostic insights related to several previously identified causative genes for these diseases. The review also provides an in-depth description of the clinicoradiologic presentations and potential underlying mitochondrial defects, aiming to enhance diagnostic abilities of radiologists in identifying primary mitochondrial diseases in their clinical practice.
Collapse
Affiliation(s)
- Sedat Giray Kandemirli
- Duke University Hospital, 2301 Erwin Rd, Durham, NC, 27710, USA.
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
| | - Khalid Al-Dasuqi
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Sidra Medical and Research Center, Doha, Qatar
| | - Bulent Aslan
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Amy Goldstein
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | |
Collapse
|
21
|
Jin S, He Y, Feng C, Yuan J, Guo Y, Guo Z, Wang X. Cellular Discrepancy of Platinum Complexes in Interfering with Mitochondrial DNA. ACS CENTRAL SCIENCE 2025; 11:393-403. [PMID: 40161961 PMCID: PMC11950849 DOI: 10.1021/acscentsci.4c01941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria are associated with cellular energy metabolism, proliferation, and mode of death. Damage to mitochondrial DNA (mtDNA) greatly affects mitochondrial function by interfering with energy production and the signaling pathway. Monofunctional trinuclear platinum complex MTPC demonstrates different actions on the mtDNA of cancerous and normal cells. It severely impairs the integrity and function of mitochondria in the human lung cancer A549 cells, such as dissipating mitochondrial membrane potential, decreasing the copy number of mtDNA, interfering in nucleoid proteins and polymerase gamma gene, reducing adenosine triphosphate (ATP), and inducing mitophagy, whereas it barely affects the mtDNA of the human kidney 2 (HK-2) cells. Moreover, MTPC promotes the release of mtDNA into the cytosol and stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, thus showing the potential to trigger antitumor immunity. MTPC displays significant cytotoxicity against A549 cells, while it exhibits weak toxicity toward HK-2 cells, therefore displaying great advantage to overcome the lingering nephrotoxicity of platinum anticancer drugs. Discrepant effects of a metal complex on mitochondria of different cells mean that targeting mitochondria has special significance in cancer therapy.
Collapse
Affiliation(s)
- Suxing Jin
- School
of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, P. R. China
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yafeng He
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Chenyao Feng
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jian Yuan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yan Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiaoyong Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
22
|
Çakmak Durmaz Ç, Langerscheidt F, Mantey I, Xia X, Zempel H. Knockdown of POLG Mimics the Neuronal Pathology of Polymerase-γ Spectrum Disorders in Human Neurons. Cells 2025; 14:480. [PMID: 40214434 PMCID: PMC11987721 DOI: 10.3390/cells14070480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Impaired function of Polymerase-γ (Pol-γ) results in impaired replication of the mitochondrial genome (mtDNA). Pathogenic mutations in the POLG gene cause dysfunctional Pol-γ and dysfunctional mitochondria and are associated with a spectrum of neurogenetic disorders referred to as POLG spectrum disorders (POLG-SDs), which are characterized by neurologic dysfunction and premature death. Pathomechanistic studies and human cell models of these diseases are scarce. SH-SY5Y cells (SHC) are an easy-to-handle and low-cost human-derived neuronal cell model commonly used in neuroscientific research. Here, we aimed to study the effect of reduced Pol-γ function using stable lentivirus-based shRNA-mediated knockdown of POLG in SHC, in both the proliferating cells and SHC-derived neurons. POLG knockdown resulted in approximately 50% reductions in POLG mRNA and protein levels in naïve SHC, mimicking the residual Pol-γ activity observed in patients with common pathogenic POLG mutations. Knockdown cells exhibited decreased mtDNA content, reduced levels of mitochondrial-encoded proteins, and altered mitochondrial morphology and distribution. Notably, while chemical induction of mtDNA depletion via ddC could be rescued by the mitochondrial biosynthesis stimulators AICAR, cilostazol and resveratrol (but not MitoQ and formoterol) in control cells, POLG-knockdown cells were resistant to mitochondrial biosynthesis-mediated induction of mtDNA increase, highlighting the specificity of the model, and pathomechanistically hinting towards inefficiency of mitochondrial stimulation without sufficient Pol-γ activity. In differentiated SHC-derived human neurons, POLG-knockdown cells showed impaired neuronal differentiation capacity, disrupted cytoskeletal organization, and abnormal perinuclear clustering of mitochondria. In sum, our model not only recapitulates key features of POLG-SDs such as impaired mtDNA content, which cannot be rescued by mitochondrial biosynthesis stimulation, but also reduced ATP production, perinuclear clustering of mitochondria and impaired neuronal differentiation. It also offers a simple, cost-effective and human (and, as such, disease-relevant) platform for investigating disease mechanisms, one with screening potential for therapeutic approaches for POLG-related mitochondrial dysfunction in human neurons.
Collapse
Affiliation(s)
- Çağla Çakmak Durmaz
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Felix Langerscheidt
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Imra Mantey
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- Institute of Human Genetics, University Hospital Magdeburg, 39120 Magdeburg, Germany
| | - Xinyu Xia
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Hans Zempel
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
23
|
Li D, Shi Y, Sun H, Yan C, Lin Y. Novel biallelic TK2 mutations cause mitochondrial DNA depletion syndrome with infantile early-onset lipid storage myopathy. Orphanet J Rare Dis 2025; 20:130. [PMID: 40098049 PMCID: PMC11912596 DOI: 10.1186/s13023-025-03639-x] [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: 11/04/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Mutations in the TK2 gene are strongly associated with mitochondrial DNA depletion syndrome (MDS), a severe condition with high mortality and poor outcomes. Although many MDS cases are reported, those linked to TK2 mutations with lipid deposition are rare. Large deletions in the TK2 gene are even rarer. METHODS We conducted whole-exome sequencing to find the gene linked to MDS, followed by genomic and structural analyses, histopathological, and functional analyses to assess the mutations' pathogenicity. Additionally, a HEK293T cell model with TK2 mutations was created to investigate the impact of large deletions on mitochondrial function. RESULTS The patient was found to have a novel compound heterozygous mutation in the TK2 gene, consisting of a large deletion spanning exons 5-10 (E5-E10 del) and a previously reported missense mutation (c.311C > A, p.Arg104His). Analysis of the patient's muscle tissue demonstrated a marked reduction in mtDNA content and a significant impairment in overall mitochondrial function. In the HEK293T cell model, the group with the deletion mutation exhibited a notable reduction in TK2 protein expression and levels of mitochondrial complex subunits when compared to the control group. Furthermore, there was an observed increase in ROS levels, a decrease in ATP production, and compromised mitochondrial respiratory chain function. Moreover, we conducted a comprehensive review of the previously reported genotypic and phenotypic spectrum of TK2 mutations in the literature. CONCLUSIONS This case report underscores the detrimental impact of large fragment deletion mutations in the TK2 gene and elucidates their role in the pathogenesis of MDS. It broadens the spectrum of known TK2 mutations and enhances our understanding of the structural and functional consequences of these mutations.
Collapse
Affiliation(s)
- Duoling Li
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, No. 107 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Yixin Shi
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, No. 107 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Hanhan Sun
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, No. 107 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Chuanzhu Yan
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, No. 107 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Yan Lin
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, No. 107 West Wenhua Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
24
|
Mabry CJ, Weindel CG, Stranahan LW, VanPortfliet JJ, Davis JR, Martinez EL, West AP, Patrick KL, Watson RO. Necrosis drives susceptibility to Mycobacterium tuberculosis in Polg D257A mutator mice. Infect Immun 2025; 93:e0032424. [PMID: 39969190 PMCID: PMC11895495 DOI: 10.1128/iai.00324-24] [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: 07/25/2024] [Accepted: 12/20/2024] [Indexed: 02/20/2025] Open
Abstract
The genetic and molecular determinants that underlie the heterogeneity of Mycobacterium tuberculosis (Mtb) infection outcomes in humans are poorly understood. Multiple lines of evidence demonstrate that mitochondrial dysfunction can exacerbate mycobacterial disease severity, and mutations in some mitochondrial genes confer susceptibility to mycobacterial infection in humans. Here, we report that mutations in mitochondria DNA (mtDNA) polymerase gamma potentiate susceptibility to Mtb infection in mice. PolgD257A mutator mtDNA mice fail to mount a protective innate immune response at an early infection time point, evidenced by high bacterial burdens, reduced M1 macrophages, and excessive neutrophil infiltration in the lungs. Immunohistochemistry reveals signs of enhanced necrosis in the lungs of Mtb-infected PolgD257A mice, and PolgD257A mutator macrophages are hypersusceptible to extrinsic triggers of necroptosis ex vivo. By assigning a role for mtDNA mutations in driving necrosis during Mtb infection, this work further highlights the requirement for mitochondrial homeostasis in mounting balanced immune responses to Mtb.
Collapse
Affiliation(s)
- C. J. Mabry
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
| | - C. G. Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
| | - L. W. Stranahan
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, Texas, USA
| | - J. J. VanPortfliet
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - J. R. Davis
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
| | - E. L. Martinez
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - A. P. West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
- The Jackson Laboratory, Bar Harbor, Maine, USA
| | - K. L. Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - R. O. Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, Texas, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
25
|
Bond ST, King EJ, Walker SM, Yang C, Liu Y, Liu KH, Zhuang A, Jurrjens AW, Fang HA, Formosa LE, Nath AP, Carmona SR, Inouye M, Duong T, Huynh K, Meikle PJ, Crawford S, Ramm G, Elahee Doomun SN, de Souza DP, Rudler DL, Calkin AC, Filipovska A, Greening DW, Henstridge DC, Drew BG. Mitochondrial damage in muscle specific PolG mutant mice activates the integrated stress response and disrupts the mitochondrial folate cycle. Nat Commun 2025; 16:2338. [PMID: 40057508 PMCID: PMC11890779 DOI: 10.1038/s41467-025-57299-3] [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: 06/23/2024] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
During mitochondrial damage, information is relayed between the mitochondria and nucleus to coordinate precise responses to preserve cellular health. One such pathway is the mitochondrial integrated stress response (mtISR), which is known to be activated by mitochondrial DNA (mtDNA) damage. However, the causal molecular signals responsible for activation of the mtISR remain mostly unknown. A gene often associated with mtDNA mutations/deletions is Polg1, which encodes the mitochondrial DNA Polymerase γ (PolG). Here, we describe an inducible, tissue specific model of PolG mutation, which in muscle specific animals leads to rapid development of mitochondrial dysfunction and muscular degeneration in male animals from ~5 months of age. Detailed molecular profiling demonstrated robust activation of the mtISR in muscles from these animals. This was accompanied by striking alterations to enzymes in the mitochondrial folate cycle that was likely driven by a specific depletion in the folate cycle metabolite 5,10 methenyl-THF, strongly implying imbalanced folate intermediates as a previously unrecognised pathology linking the mtISR and mitochondrial disease.
Collapse
Affiliation(s)
- Simon T Bond
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Emily J King
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Shannen M Walker
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | | | - Yingying Liu
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Kevin H Liu
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Aowen Zhuang
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Aaron W Jurrjens
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Haoyun A Fang
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Luke E Formosa
- Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Artika P Nath
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | | | | | - Thy Duong
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Kevin Huynh
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Peter J Meikle
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Simon Crawford
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Australia
| | - Georg Ramm
- Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Australia
| | | | | | - Danielle L Rudler
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Western Australia, Nedlands, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Western Australia, Nedlands, Australia
| | - Anna C Calkin
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Aleksandra Filipovska
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Western Australia, Nedlands, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Western Australia, Nedlands, Australia
| | - David W Greening
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Darren C Henstridge
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Brian G Drew
- Baker Heart & Diabetes Institute, Melbourne, Australia.
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia.
- School of Translational Medicine, Monash University, Melbourne, Australia.
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia.
| |
Collapse
|
26
|
Deng H, Zhang Q, Yi J, Yuan L. Unraveling ptosis: A comprehensive review of clinical manifestations, genetics, and treatment. Prog Retin Eye Res 2025; 105:101327. [PMID: 39725023 DOI: 10.1016/j.preteyeres.2024.101327] [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: 09/05/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Ptosis is defined as an abnormally low-lying upper eyelid margin on the primary gaze, generally resulting from a congenital or acquired abnormality of the nerves or muscles that control the eyelid. Ptosis can occur alone or concurrently as an ocular or systemic syndrome, and the prevalence of ptosis varies among different countries and populations. Isolated ptosis typically causes aesthetic problems in patients and can lead to functional ophthalmic problems in severe cases. In individuals with syndromic ptosis, ptosis can be a warning of serious medical problems. There are different approaches to classification, depending on the onset time or the etiology of ptosis, and the clinical characteristics of congenital and acquired ptosis also differ. Pedigree and genetic analysis have demonstrated that hereditary ptosis is clinically heterogeneous, with incomplete concordance and variable expressivity. A number of genetic loci and genes responsible for hereditary isolated and syndromic ptosis have been reported. Optimal surgical timing and proper method are truly critical for avoiding the risk of potentially severe outcomes from ptosis and minimizing surgical complications, which are challenging as the pathogenesis is still indistinct and the anatomy is complex. This review provides a comprehensive review of ptosis, by summarizing the clinical manifestations, classification, diagnosis, genetics, treatment, and prognosis, as well as the bound anatomy of upper eyelid.
Collapse
Affiliation(s)
- Hao Deng
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Research Center of Medical Experimental Technology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Disease Genome Research Center, Central South University, Changsha, 410013, China; Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Qianling Zhang
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Junhui Yi
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Lamei Yuan
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Research Center of Medical Experimental Technology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Disease Genome Research Center, Central South University, Changsha, 410013, China.
| |
Collapse
|
27
|
Ivaniuk A, Anselm IA, Bowen A, Cohen BH, Eminoglu FT, Estrella J, Gallagher RC, Ganetzky RD, Gannon J, Gorman GS, Greene C, Gropman AL, Haas RH, Hirano M, Kapoor S, Karaa A, Koenig MK, Kornblum C, Kose E, Larson A, Lichter-Konecki U, Lopriore P, Mancuso M, McFarland R, Moe AM, Morava E, Ng YS, Saneto RP, Scaglia F, Sue CM, Tarnopolsky M, Walker MA, Parikh S, as the Hong Kong Mitochondrial Diseases Interest Group, Cheuk-Wing F, Wong TS, Belaramani K, Chan CK, Chan WK, Chan WLL, Cheung HW, Cheung KY, Chang SK, Cheung SN, Cheung TF, Cheung YF, Chong SCJ, Chow CKJ, Chung HYB, Fan SYF, Fok WMJ, Fong KW, Fung THS, Hui KF, Hui TH, Hui J, Ko CH, Kwan MC, Kwok MKA, Kwok SSJ, Lai MS, Lam YO, Lam CW, Lau MC, Law CYE, Law HF, Lee WC, Hencher Lee HC, Leung KH, Leung KY, Li SH, Ling TKJ, Liu KTT, Lo FM, Lui C, Luk CO, Luk HM, Ma CK, Ma K, Ma KH, Mew YN, Mo A, Hg SF, Poon WKG, Sheng B, Szeto CLC, Tai SM, Tang JL, Tse CTA, Tsung LYL, Wong HMJ, Wong WYW, Wong KK, Wong SNS, Wong CNV, Wong WSS, Wong CKF, Wu SP, Wu HFJ, Yau MM, et alIvaniuk A, Anselm IA, Bowen A, Cohen BH, Eminoglu FT, Estrella J, Gallagher RC, Ganetzky RD, Gannon J, Gorman GS, Greene C, Gropman AL, Haas RH, Hirano M, Kapoor S, Karaa A, Koenig MK, Kornblum C, Kose E, Larson A, Lichter-Konecki U, Lopriore P, Mancuso M, McFarland R, Moe AM, Morava E, Ng YS, Saneto RP, Scaglia F, Sue CM, Tarnopolsky M, Walker MA, Parikh S, as the Hong Kong Mitochondrial Diseases Interest Group, Cheuk-Wing F, Wong TS, Belaramani K, Chan CK, Chan WK, Chan WLL, Cheung HW, Cheung KY, Chang SK, Cheung SN, Cheung TF, Cheung YF, Chong SCJ, Chow CKJ, Chung HYB, Fan SYF, Fok WMJ, Fong KW, Fung THS, Hui KF, Hui TH, Hui J, Ko CH, Kwan MC, Kwok MKA, Kwok SSJ, Lai MS, Lam YO, Lam CW, Lau MC, Law CYE, Law HF, Lee WC, Hencher Lee HC, Leung KH, Leung KY, Li SH, Ling TKJ, Liu KTT, Lo FM, Lui C, Luk CO, Luk HM, Ma CK, Ma K, Ma KH, Mew YN, Mo A, Hg SF, Poon WKG, Sheng B, Szeto CLC, Tai SM, Tang JL, Tse CTA, Tsung LYL, Wong HMJ, Wong WYW, Wong KK, Wong SNS, Wong CNV, Wong WSS, Wong CKF, Wu SP, Wu HFJ, Yau MM, Yau KCE, Yeung WL, Yeung HMJ, Yip KKE, Wu HJ, Young PHT, Yuan G, Yuen YPL, Yuen CL. Characterization of Factors Associated With Death in Deceased Patients With Mitochondrial Disorders: A Multicenter Cross-Sectional Survey. Neurology 2025; 104:e209779. [PMID: 39883904 PMCID: PMC11781783 DOI: 10.1212/wnl.0000000000209779] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 11/14/2024] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Mitochondrial disorders are multiorgan disorders resulting in significant morbidity and mortality. We aimed to characterize death-associated factors in an international cohort of deceased individuals with mitochondrial disorders. METHODS This cross-sectional multicenter observational study used data provided by 26 mitochondrial disease centers from 8 countries from January 2022 to March 2023. Individuals with genetically confirmed mitochondrial disorders were included, along with patients with clinically or genetically diagnosed Leigh syndrome. Collected data included demographic and genetic diagnosis variables, clinical phenotype, involvement of organs and systems, conditions leading to death, and supportive care. We defined pediatric and adult groups based on age at death before or after 18 years, respectively. We used Kruskal-Wallis with post hoc Dunn test with Bonferroni correction and Fisher exact test for comparisons, Spearman rank test for correlations, and multiple linear regression for multivariable analysis. RESULTS Data from 330 deceased individuals with mitochondrial disorders (191 [57.9%] pediatric) were analyzed. The shortest survival times were observed in hepatocerebral syndrome (median 0.3, interquartile range [IQR] 0.2-0.6 years) and mitochondrial cardiomyopathy (median 0.3, IQR 0.2-5.2 years) and the longest in chronic progressive external ophthalmoplegia plus (median 26.5, IQR 22.8-40.2 years) and sensory ataxic neuropathy, dysarthria, and ophthalmoparesis (median 21.0, IQR 13.8-28.5 years). Respiratory failure and pulmonary infections were the most common conditions associated with death (52/330, 15.7% and 46/330, 13.9%, respectively). Noninvasive ventilation was required more often in children (57/191, 29.8%) than adults (12/139, 8.6%, p < 0.001), as was nasogastric or gastric tube (131/191, 68.6% in children and 39/139, 28.1% in adults, p < 0.001). On multivariate analysis, individuals with movement disorders and nuclear gene involvement had increased odds of any respiratory support use (OR 2.42 (95% CI 1.17-5.22) and OR 2.39 (95% CI 1.16-5.07), respectively). DISCUSSION This international collaboration highlights the importance of respiratory care and infection management and provides a reference for prognostication across different mitochondrial disorders.
Collapse
Affiliation(s)
| | | | - Aaron Bowen
- Department of Neurology, Boston Children's Hospital, MA
| | - Bruce H. Cohen
- Department of Pediatrics, Neurodevelopmental Science Center, Children's Hospital Medical Center of Akron, OH
| | - Fatma Tuba Eminoglu
- Ankara University Faculty of Medicine, Department of Pediatric Metabolism and Rare Diseases Application and Research Center, Turkey;
| | - Jane Estrella
- School of Medicine, Macarthur Clinical School, Western Sydney University, New South Wales, Australia;
| | | | - Rebecca D. Ganetzky
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine
| | | | - Grainne S. Gorman
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University; NHS Highly Specialized Service for Rare Mitochondrial Disorders, Newcastle upon Tyne, United Kingdom
| | - Carol Greene
- Department of Pediatrics, University of Maryland Medical Center, Baltimore
| | - Andrea L. Gropman
- Division of Neurogenetics and Developments Pediatrics, Children's National Health System, Washington, DC
| | - Richard H. Haas
- Department of Neurosciences and Pediatrics, UCSD Medical Center and Rady Children's Hospital San Diego, La Jolla, CA
| | - Michio Hirano
- Department of Neurology, Columbia University Medical Center, New York, NY
| | - Seema Kapoor
- Department of Pediatrics, Maulana Azad Medical College and Lok Nayak Hospital, New Delhi, India;
| | - Amel Karaa
- Pediatric Department, Clinical Genetics Division, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mary Kay Koenig
- Department of Pediatrics, Division of Child and Adolescent Neurology, The University of Texas McGovern Medical School, Houston
| | - Cornelia Kornblum
- Department of Neurology, Neuromuscular Disease Section; Centre for Rare Diseases, University Hospital Bonn, Germany;
| | - Engin Kose
- Ankara University Faculty of Medicine, Department of Pediatric Metabolism and Rare Diseases Application and Research Center, Turkey;
| | - Austin Larson
- Section of Genetics, Department of Pediatrics, University of Colorado Denver and Children's Hospital Colorado, Aurora
| | - Uta Lichter-Konecki
- Division Medical Genetics, Department of Pediatrics, University of Pittsburgh, School of Medicine, Center for Rare Disease Therapy, UPMC Children's Hospital of Pittsburgh, PA
| | - Piervito Lopriore
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Italy;
| | - Michelangelo Mancuso
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Italy;
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University; NHS Highly Specialized Service for Rare Mitochondrial Disorders, Newcastle upon Tyne, United Kingdom
| | - Aye Myat Moe
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University; NHS Highly Specialized Service for Rare Mitochondrial Disorders, Newcastle upon Tyne, United Kingdom
| | - Eva Morava
- Department of Clinical Genomics and Laboratory of Medical Pathology, Mayo Clinic, Rochester, MN
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University; NHS Highly Specialized Service for Rare Mitochondrial Disorders, Newcastle upon Tyne, United Kingdom
| | - Russell P. Saneto
- Neuroscience Institute, Center for Integrated Brain Research, Department of Neurology and Division of Pediatric Neurology, Seattle Children's Hospital and University of Washington
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine; Texas Children's Hospital; Joint BCM-CUHK Center of Medical Genetics, Prince of Wales Hospital, Chinese University of Hong Kong, China
| | - Carolyn M. Sue
- Department of Neurogenetics, Kolling Institute, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia;
| | - Mark Tarnopolsky
- Department of Pediatrics and Medicine, McMaster Children's Hospital, Hamilton, Ontario, Canada;
| | | | - Sumit Parikh
- Mitochondrial Medicine Center, Department of Neurology, Center for Child Neurology, Cleveland Clinic Children's Hospital, OH
| | - as the Hong Kong Mitochondrial Diseases Interest Group
- Epilepsy Center, Cleveland Clinic, OH
- Department of Neurology, Boston Children's Hospital, MA
- Department of Pediatrics, Neurodevelopmental Science Center, Children's Hospital Medical Center of Akron, OH
- Ankara University Faculty of Medicine, Department of Pediatric Metabolism and Rare Diseases Application and Research Center, Turkey;
- School of Medicine, Macarthur Clinical School, Western Sydney University, New South Wales, Australia;
- Institute for Human Genetics, University of California, San Francisco, CA
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine
- Division of Genetics, Children's Mercy Kansas City, MO
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University; NHS Highly Specialized Service for Rare Mitochondrial Disorders, Newcastle upon Tyne, United Kingdom
- Department of Pediatrics, University of Maryland Medical Center, Baltimore
- Division of Neurogenetics and Developments Pediatrics, Children's National Health System, Washington, DC
- Department of Neurosciences and Pediatrics, UCSD Medical Center and Rady Children's Hospital San Diego, La Jolla, CA
- Department of Neurology, Columbia University Medical Center, New York, NY
- Department of Pediatrics, Maulana Azad Medical College and Lok Nayak Hospital, New Delhi, India;
- Pediatric Department, Clinical Genetics Division, Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Pediatrics, Division of Child and Adolescent Neurology, The University of Texas McGovern Medical School, Houston
- Department of Neurology, Neuromuscular Disease Section; Centre for Rare Diseases, University Hospital Bonn, Germany;
- Section of Genetics, Department of Pediatrics, University of Colorado Denver and Children's Hospital Colorado, Aurora
- Division Medical Genetics, Department of Pediatrics, University of Pittsburgh, School of Medicine, Center for Rare Disease Therapy, UPMC Children's Hospital of Pittsburgh, PA
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Italy;
- Department of Clinical Genomics and Laboratory of Medical Pathology, Mayo Clinic, Rochester, MN
- Neuroscience Institute, Center for Integrated Brain Research, Department of Neurology and Division of Pediatric Neurology, Seattle Children's Hospital and University of Washington
- Department of Molecular and Human Genetics, Baylor College of Medicine; Texas Children's Hospital; Joint BCM-CUHK Center of Medical Genetics, Prince of Wales Hospital, Chinese University of Hong Kong, China
- Department of Neurogenetics, Kolling Institute, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia;
- Department of Pediatrics and Medicine, McMaster Children's Hospital, Hamilton, Ontario, Canada;
- Department of Neurology, Massachusetts General Hospital, Boston
- Mitochondrial Medicine Center, Department of Neurology, Center for Child Neurology, Cleveland Clinic Children's Hospital, OH
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital;
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China;
- Department of Medicine, Queen Mary Hospital, Hong Kong, China;
- Department of Medicine and Geriatrics, Kwong Wah Hospital, Hong Kong, China;
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, China;
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, Queen Elizabeth Hospital, Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong; Hong Kong Genome Institute, China;
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong, China;
- Department of Medicine, Yan Chai Hospital, Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, Kwong Wah Hospital, Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, Caritas Medical Centre, Hong Kong, China;
- Department of Chemical Pathology, Prince of Wales Hospital, Hong Kong, China;
- Department of Medicine, North District Hospital, Hong Kong, China;
- Department of Pathology, The University of Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, United Christian Hospital, Hong Kong, China;
- Department of Chemical Pathology, Queen Mary Hospital, Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China;
- Department of Chemical Pathology, Princess Margaret Hospital, Hong Kong, China;
- Department of Health, Clinical Genetic Service, Hong Kong, China;
- Department of Medicine, Tseung Kwan O Hospital, Hong Kong, China;
- Clinical Genetics Service Unit, Hong Kong Children's Hospital, China;
- Department of Paediatrics and Adolescent Medicine, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong, China;
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China;
- Department of Medicine and Geriatrics, Caritas Medical Centre, Hong Kong, China;
- Department of Pathology, Queen Elizabeth Hospital, Hong Kong, China;
- Department of Paediatrics and Adolescent Medicine, Tseung Kwan O Hospital, Hong Kong, China;
- Department of Medicine and Geriatrics, Ruttonjee and Tang Shiu Kin Hospitals, Hong Kong, China;
- Department of Chemical Pathology, Hong Kong Children's Hospital, China; and
- Department of Medicine and Geriatrics, Tuen Mun Hospital, Hong Kong, People's Republic of China
| | - Fung Cheuk-Wing
- Department of Pediatrics and Adolescent Medicine, Hong Kong Children's Hospital;
| | - Tsz-Sum Wong
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China;
| | - Kiran Belaramani
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
| | - Chun-Kong Chan
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Wing-Ki Chan
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China;
| | - Wai-Lun Larry Chan
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China;
| | - Hon-Wing Cheung
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Ka-Yin Cheung
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Shek-Kwan Chang
- Department of Medicine, Queen Mary Hospital, Hong Kong, China;
| | - Sing-Ngai Cheung
- Department of Medicine and Geriatrics, Kwong Wah Hospital, Hong Kong, China;
| | - Tsz-Fung Cheung
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Yuk-Fai Cheung
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, China;
| | | | - Chi-Kwan Jasmine Chow
- Department of Paediatrics and Adolescent Medicine, Queen Elizabeth Hospital, Hong Kong, China;
| | - Hon-Yin B. Chung
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong; Hong Kong Genome Institute, China;
| | - Sin-Ying Florence Fan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong, China;
| | | | - Ka-Wing Fong
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, China;
| | - Tsui-Hang Sharon Fung
- Department of Paediatrics and Adolescent Medicine, Kwong Wah Hospital, Hong Kong, China;
| | - Kwok-Fai Hui
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Ting-Hin Hui
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Joannie Hui
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
| | - Chun Hung Ko
- Department of Paediatrics and Adolescent Medicine, Caritas Medical Centre, Hong Kong, China;
| | - Min-Chung Kwan
- Department of Medicine and Geriatrics, Kwong Wah Hospital, Hong Kong, China;
| | - Mei-Kwan Anne Kwok
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
| | | | - Moon-Sing Lai
- Department of Medicine, North District Hospital, Hong Kong, China;
| | - Yau-On Lam
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Ching-Wan Lam
- Department of Pathology, The University of Hong Kong, China;
| | - Ming-Chung Lau
- Department of Paediatrics and Adolescent Medicine, United Christian Hospital, Hong Kong, China;
| | - Chun-Yiu Eric Law
- Department of Chemical Pathology, Queen Mary Hospital, Hong Kong, China;
| | - Hiu-Fung Law
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
| | - Wing-Cheong Lee
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China;
| | | | - Kin-Hang Leung
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, China;
| | - Kit-Yan Leung
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China;
| | - Siu-Hung Li
- Department of Medicine, North District Hospital, Hong Kong, China;
| | - Tsz-Ki Jacky Ling
- Department of Chemical Pathology, Queen Mary Hospital, Hong Kong, China;
| | - Kam-Tim Timothy Liu
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China;
| | - Fai-Man Lo
- Department of Health, Clinical Genetic Service, Hong Kong, China;
| | - Colin Lui
- Department of Medicine, Tseung Kwan O Hospital, Hong Kong, China;
| | - Ching-On Luk
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, China;
| | - Ho-Ming Luk
- Clinical Genetics Service Unit, Hong Kong Children's Hospital, China;
| | - Che-Kwan Ma
- Department of Paediatrics and Adolescent Medicine, United Christian Hospital, Hong Kong, China;
| | - Karen Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong, China;
| | - Kam-Hung Ma
- Department of Paediatrics and Adolescent Medicine, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China;
| | - Yuen-Ni Mew
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | - Alex Mo
- Department of Paediatrics and Adolescent Medicine, Kwong Wah Hospital, Hong Kong, China;
| | - Sui-Fun Hg
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China;
| | - Wing-Kit Grace Poon
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong, China;
| | - Bun Sheng
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China;
| | | | - Shuk-Mui Tai
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China;
| | - Jing-Liang Tang
- Department of Medicine and Geriatrics, United Christian Hospital, Hong Kong, China;
| | | | - Li-Yan Lilian Tsung
- Department of Paediatrics and Adolescent Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China;
| | - Ho-Ming June Wong
- Department of Medicine and Geriatrics, Caritas Medical Centre, Hong Kong, China;
| | - Wing-Yin Winnie Wong
- Department of Medicine and Geriatrics, Caritas Medical Centre, Hong Kong, China;
| | - Kwok-Kui Wong
- Department of Medicine, Yan Chai Hospital, Hong Kong, China;
| | - Suet-Na Sheila Wong
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
| | - Chun-Nei Virginia Wong
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong; Hong Kong Genome Institute, China;
| | | | - Chi-Kin Felix Wong
- Department of Chemical Pathology, Queen Mary Hospital, Hong Kong, China;
| | - Shun-Ping Wu
- Department of Paediatrics and Adolescent Medicine, Queen Elizabeth Hospital, Hong Kong, China;
| | - Hiu-Fung Jerome Wu
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China;
| | - Man-Mut Yau
- Department of Paediatrics and Adolescent Medicine, Tseung Kwan O Hospital, Hong Kong, China;
| | - Kin-Cheong Eric Yau
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China;
| | - Wai-Lan Yeung
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
| | - Hon-Ming Jonas Yeung
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital, Hong Kong, China;
| | - Kin-Keung Edwin Yip
- Department of Medicine and Geriatrics, Ruttonjee and Tang Shiu Kin Hospitals, Hong Kong, China;
| | - Hui-Jun Wu
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, China;
| | - Pui-Hong Terence Young
- Department of Medicine and Geriatrics, Ruttonjee and Tang Shiu Kin Hospitals, Hong Kong, China;
| | - Gao Yuan
- Department of Medicine, Queen Mary Hospital, Hong Kong, China;
| | - Yuet-Ping Liz Yuen
- Department of Chemical Pathology, Hong Kong Children's Hospital, China; and
| | - Chi-Lap Yuen
- Department of Medicine and Geriatrics, Tuen Mun Hospital, Hong Kong, People's Republic of China
| |
Collapse
|
28
|
Finn LS, Goldstein A, Hedrick HL. Mitochondrial Neurogastrointestinal Encephalomyopathy (MNGIE) Phenotype Associated With Unique Compound Heterozygous POLG Variants: Case Presentation and Review of the Literature. Pediatr Dev Pathol 2025:10935266251321317. [PMID: 39982139 DOI: 10.1177/10935266251321317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
We report a teenage patient with a delayed diagnosis of compound heterozygous POLG pathogenic variants [(POLG c. 1943 C>G, p.P648R) and (POLG c. 679 C>T, p.R227W)] who presented with fatigue and neuropathy, as well as long standing malnutrition and cachexia, erroneously attributed to an eating disorder. She experienced multiple bowel perforations and pathologic examination revealed jejunal diverticula and features of visceral neuromyopathy. In addition to ganglion cell mega-mitochondrial inclusions, there were multiple foci of interrupted muscularis mucosae, an alteration not previously recognized in the intestines of patients with primary mitochondrial disorders. We provide a detailed account of the gastrointestinal pathologic findings in this patient and compare with prior cases of Mitochondrial Neurogastrointestinal Encephalomyopathy (MNGIE) phenotypes.
Collapse
Affiliation(s)
- Laura S Finn
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Goldstein
- Division of Human Genetics, Mitochondrial Medicine Frontier Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Holly L Hedrick
- Division of General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
29
|
Rothwell S, Ng I, Shalchy-Tabrizi S, Kalinowski P, Taha OM, Paris I, Baniqued A, Lin L, Mezei MM, Lehman A, Julian LM, Poburko D. Loss-of-function mitochondrial DNA polymerase gamma variants cause vascular smooth muscle cells to secrete a diffusible mitogenic factor. Front Physiol 2025; 15:1488248. [PMID: 40034369 PMCID: PMC11873068 DOI: 10.3389/fphys.2024.1488248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/26/2024] [Indexed: 03/05/2025] Open
Abstract
Introduction Mitochondrial dysfunction promotes vascular aging and disease through diverse mechanisms beyond metabolic supply, including calcium and radical signaling and inflammation. Mitochondrial DNA (mtDNA) replication by the POLG-encoded mitochondrial DNA polymerase (POLG) is critical for mitochondrial health. Loss-of-function POLG variants are associated with a predisposition to hypertension. We hypothesized that impaired POLG, through reduced mtDNA copy number or other mechanisms, would promote smooth muscle hypertrophy or hyperplasia that drives vascular remodeling associated with hypertension. Methods We characterized the effect of over-expressing POLG variants that were previously observed in a cohort of hypertensive patients (p.Tyr955Cys, p.Arg964Cys, p.Asn1098Ile, and p.Arg1138Cys) in A7r5 cells. Results AlphaFold modeling of the POLG holoenzyme complexed with DNA predicted changes in the catalytic site in the p.Tyr955Cys and p.Asn1098Ile variants, while p.Arg964Cys and p.Arg1138Cys showed minimal effects. The POLG variants reduced mtDNA copy number, assessed by immunofluorescence and droplet digital PCR, by up to 27% in the order p.Tyr955Cys > p.Arg964Cys > p.Asn1098Ile > p.Arg1138Cys relative to wild-type-transfected cultures. Loss of mtDNA was reduced in cultures grown in low serum and glucose media, but the cell density was increased in the same rank order in both 10% serum and 1% serum. POLG constructs contained a Myc epitope, the counterstaining for which showed that the mtDNA copy number was reduced in both transfected cells and untransfected neighbors. Live-cell imaging of mitochondrial membrane potential with TMRM and radical oxygen species production with MitoSOX showed little effect of the POLG variants. POLG variants had little effect on oxygen consumption, assessed by Seahorse assay. Live-cell imaging growth analyses again showed increased growth in A7r5 cells transfected with p.Tyr955Cys but a decreased growth with p.Arg1138Cys, while p.Tyr955Cys increased growth of HeLa cells. Conditioned media from HeLa cells transfected with POLG variants reduced doubling times in naïve cultures. Pharmacologically, wedelolactone and MitoTEMPOL, but not indomethacin or PD98059, suppressed the mitogenic effects of p.Tyr955Cys and p.Arg964Cys in A7r5 cells. Discussion We conclude that POLG dysfunction induces secretion of a mitogenic signal from A7r5 and HeLa cells even when changes in mtDNA copy number are below the limit of detection. Such mitogenic stimulation could stimulate hypertrophic remodeling that could contribute to drug-resistant hypertension in patient populations with loss-of-function POLG variants.
Collapse
Affiliation(s)
- Samantha Rothwell
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Irvin Ng
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | | | - Pola Kalinowski
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Omnia M. Taha
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Italia Paris
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Angelica Baniqued
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa Lin
- Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology Development and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
| | - Michelle M. Mezei
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada
- Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Lisa M. Julian
- Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology Development and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
| | - Damon Poburko
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
30
|
Yoshihara R, Shimakura Y, Kitamura S, Satoh K, Sato M, Aono T, Akiyama Y, Hatakeyama S, Tanaka S. A mutation in DNA polymerase γ harbors a shortened lifespan and high sensitivity to mutagens in the filamentous fungus Neurospora crassa. Genetics 2025; 229:iyae201. [PMID: 39611774 DOI: 10.1093/genetics/iyae201] [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: 07/03/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
Hyphal elongation is the vegetative growth of filamentous fungi, and many species continuously elongate their hyphal tips over long periods. The details of the mechanisms for maintaining continuous growth are not yet clear. A novel short lifespan mutant of N. crassa that ceases hyphal elongation early was screened and analyzed to better understand the mechanisms for maintaining hyphal elongation in filamentous fungi. The mutant strain also exhibited high sensitivity to mutagens such as hydroxyurea and ultraviolet radiation. Based on these observations, we named the novel mutant "mutagen sensitive and short lifespan 1 (ms1)." The mutation responsible for the short lifespan and mutagen sensitivity in the ms1 strain was identified in DNA polymerase γ (mip-1:NCU00276). This mutation changed the amino acid at position 814 in the polymerase domain from leucine to arginine (MIP-1 L814R). A dosage analysis by next-generation sequencing reads suggested that mitochondrial DNA (mtDNA) sequences are decreased nonuniformly throughout the genome of the ms1 strain. This observation was confirmed by quantitative PCR for 3 representative loci and restriction fragment length polymorphisms in purified mtDNA. Direct repeat-mediated deletions, which had been reported previously, were not detected in the mitochondrial genome by our whole-genome sequencing analysis. These results imply the presence of novel mechanisms to induce the nonuniform decrease in the mitochondrial genome by DNA polymerase γ mutation. Some potential reasons for the nonuniform distribution of the mitochondrial genome are discussed in relation to the molecular functions of DNA polymerase γ.
Collapse
Affiliation(s)
- Ryouhei Yoshihara
- Graduate School of Science & Engineering, Saitama University, Shimo-Ohkubo 255, Sakura-ku, Saitama, Saitama 338-8570, Japan
| | - Yuzuki Shimakura
- Graduate School of Science & Engineering, Saitama University, Shimo-Ohkubo 255, Sakura-ku, Saitama, Saitama 338-8570, Japan
| | - Satoshi Kitamura
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Takasaki 370-1292, Japan
| | - Katsuya Satoh
- Takasaki Institute for Advanced Quantum Science, National Institutes for Quantum Science and Technology (QST), Takasaki 370-1292, Japan
| | - Manami Sato
- Graduate School of Science & Engineering, Saitama University, Shimo-Ohkubo 255, Sakura-ku, Saitama, Saitama 338-8570, Japan
| | - Taketo Aono
- Graduate School of Science & Engineering, Saitama University, Shimo-Ohkubo 255, Sakura-ku, Saitama, Saitama 338-8570, Japan
| | - Yu Akiyama
- Graduate School of Science & Engineering, Saitama University, Shimo-Ohkubo 255, Sakura-ku, Saitama, Saitama 338-8570, Japan
| | - Shin Hatakeyama
- Graduate School of Science & Engineering, Saitama University, Shimo-Ohkubo 255, Sakura-ku, Saitama, Saitama 338-8570, Japan
| | - Shuuitsu Tanaka
- Graduate School of Science & Engineering, Saitama University, Shimo-Ohkubo 255, Sakura-ku, Saitama, Saitama 338-8570, Japan
| |
Collapse
|
31
|
Magielski JH, Ruggiero SM, Xian J, Parthasarathy S, Galer PD, Ganesan S, Back A, McKee JL, McSalley I, Gonzalez AK, Morgan A, Donaher J, Helbig I. The clinical and genetic spectrum of paediatric speech and language disorders. Brain 2025; 148:663-674. [PMID: 39412438 PMCID: PMC11788197 DOI: 10.1093/brain/awae264] [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: 03/29/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 10/23/2024] Open
Abstract
Speech and language disorders are known to have a substantial genetic contribution. Although frequently examined as components of other conditions, research on the genetic basis of linguistic differences as separate phenotypic subgroups has been limited so far. Here, we performed an in-depth characterization of speech and language disorders in 52 143 individuals, reconstructing clinical histories using a large-scale data-mining approach of the electronic medical records from an entire large paediatric healthcare network. The reported frequency of these disorders was the highest between 2 and 5 years old and spanned a spectrum of 26 broad speech and language diagnoses. We used natural language processing to assess the degree to which clinical diagnoses in full-text notes were reflected in ICD-10 diagnosis codes. We found that aphasia and speech apraxia could be retrieved easily through ICD-10 diagnosis codes, whereas stuttering as a speech phenotype was coded in only 12% of individuals through appropriate ICD-10 codes. We found significant comorbidity of speech and language disorders in neurodevelopmental conditions (30.31%) and, to a lesser degree, with epilepsies (6.07%) and movement disorders (2.05%). The most common genetic disorders retrievable in our analysis of electronic medical records were STXBP1 (n = 21), PTEN (n = 20) and CACNA1A (n = 18). When assessing associations of genetic diagnoses with specific linguistic phenotypes, we observed associations of STXBP1 and aphasia (P = 8.57 × 10-7, 95% confidence interval = 18.62-130.39) and MYO7A with speech and language development delay attributable to hearing loss (P = 1.24 × 10-5, 95% confidence interval = 17.46-infinity). Finally, in a sub-cohort of 726 individuals with whole-exome sequencing data, we identified an enrichment of rare variants in neuronal receptor pathways, in addition to associations of UQCRC1 and KIF17 with expressive aphasia, MROH8 and BCHE with poor speech, and USP37, SLC22A9 and UMODL1 with aphasia. In summary, our study outlines the landscape of paediatric speech and language disorders, confirming the phenotypic complexity of linguistic traits and novel genotype-phenotype associations. Subgroups of paediatric speech and language disorders differ significantly with respect to the composition of monogenic aetiologies.
Collapse
Affiliation(s)
- Jan H Magielski
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
| | - Sarah M Ruggiero
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Julie Xian
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
| | - Shridhar Parthasarathy
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
| | - Peter D Galer
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shiva Ganesan
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
| | - Amanda Back
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jillian L McKee
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ian McSalley
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
| | - Alexander K Gonzalez
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
| | - Angela Morgan
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Department of Audiology and Speech Pathology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Joseph Donaher
- Center for Childhood Communication, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ingo Helbig
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19146, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
32
|
Flaherty S, Song L, Albuquerque B, Rinaldi A, Piper M, Shanthappa D, Chen X, Stansfield J, Asano S, Pashos E, Ross T, Jagarlapudi S, Sheikh A, Zhang B, Wu Z. GDF15 Neutralization Ameliorates Muscle Atrophy and Exercise Intolerance in a Mouse Model of Mitochondrial Myopathy. J Cachexia Sarcopenia Muscle 2025; 16:e13715. [PMID: 39976232 PMCID: PMC11840706 DOI: 10.1002/jcsm.13715] [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: 12/21/2023] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Primary mitochondrial myopathies (PMMs) are disorders caused by mutations in genes encoding mitochondrial proteins and proteins involved in mitochondrial function. PMMs are characterized by loss of muscle mass and strength as well as impaired exercise capacity. Growth/Differentiation Factor 15 (GDF15) was reported to be highly elevated in PMMs and cancer cachexia. Previous studies have shown that GDF15 neutralization is effective in improving skeletal muscle mass and function in cancer cachexia. It remains to be determined if the inhibition of GDF15 could be beneficial for PMMs. The purpose of the present study is to assess whether treatment with a GDF15 neutralizing antibody can alleviate muscle atrophy and physical performance impairment in a mouse model of PMM. METHODS The effects of GDF15 neutralization on PMM were assessed using PolgD257A/D257A (POLG) mice. These mice express a proofreading-deficient version of the mitochondrial DNA polymerase gamma, leading to an increased rate of mutations in mitochondrial DNA (mtDNA). These animals display increased circulating GDF15 levels, reduced muscle mass and function, exercise intolerance, and premature aging. Starting at 9 months of age, the mice were treated with an anti-GDF15 antibody (mAB2) once per week for 12 weeks. Body weight, food intake, body composition, and muscle mass were assessed. Muscle function and exercise capacity were evaluated using in vivo concentric max force stimulation assays, forced treadmill running and voluntary home-cage wheel running. Mechanistic investigations were performed via muscle histology, bulk transcriptomic analysis, RT-qPCR and western blotting. RESULTS Anti-GDF15 antibody treatment ameliorated the metabolic phenotypes of the POLG animals, improving body weight (+13% ± 8%, p < 0.0001), lean mass (+13% ± 15%, p < 0.001) and muscle mass (+35% ± 24%, p < 0.001). Additionally, the treatment improved skeletal muscle max force production (+35% ± 43%, p < 0.001) and exercise performance, including treadmill (+40% ± 29%, p < 0.05) and voluntary wheel running (+320% ± 19%, p < 0.05). Mechanistically, the beneficial effects of GDF15 neutralization are linked to the reversal of the transcriptional dysregulation in genes involved in autophagy and proteasome signalling. The treatment also appears to dampen glucocorticoid signalling by suppressing circulating corticosterone levels in the POLG animals. CONCLUSIONS Our findings highlight the potential of GDF15 neutralization with a monoclonal antibody as a therapeutic avenue to enhance physical performance and mitigate adverse clinical outcomes in patients with PMM.
Collapse
Affiliation(s)
- Stephen E. Flaherty
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Obesity and ComplicationsEli LillyBostonMassachusettsUSA
| | - LouJin Song
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bina Albuquerque
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Anthony Rinaldi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Program Mamager, Preclinical Sciences, ToxicologyVertex PharmaceuticalsBostonMassachusettsUSA
| | - Mary Piper
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | | | - Xian Chen
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - John Stansfield
- Biostatistics, Early Clinical DevelopmentPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Shoh Asano
- Inflammation and Immunology Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Evanthia Pashos
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Trenton Thomas Ross
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Srinath Jagarlapudi
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
| | - Abdul Sheikh
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Bei Zhang
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| | - Zhidan Wu
- Internal Medicine Research UnitPfizer Worldwide Research, Development & MedicalCambridgeMassachusettsUSA
- Diabetes, Obesity and MASH, Global Drug DiscoveryNovo NordiskLexingtonMassachusettsUSA
| |
Collapse
|
33
|
Carvalho Araújo L, Lopes Tenório de Cerqueira IC, de Aguiar Coelho Silva Madeiro B. Teaching NeuroImage: Notable MRI Findings in Sensory Ataxia Neuropathy, Dysarthria, and Ophthalmoplegia (SANDO), a POLG-Related Disorder. Neurology 2025; 104:e210227. [PMID: 39724533 DOI: 10.1212/wnl.0000000000210227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/28/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- Luziany Carvalho Araújo
- Department of Radiology, Hospital das Clínicas, Universidade Federal de Pernambuco (UFPE) - EBSERH, Recife, Brazil; and
| | | | | |
Collapse
|
34
|
VanPortfliet JJ, Lei Y, Ramanathan M, Martinez CG, Wong J, Stodola TJ, Hoffmann BR, Pflug K, Sitcheran R, Kneeland SC, Murray SA, McGuire PJ, Cannon CL, West AP. Caspase-11 drives macrophage hyperinflammation in models of Polg-related mitochondrial disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.11.593693. [PMID: 38798587 PMCID: PMC11118447 DOI: 10.1101/2024.05.11.593693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mitochondrial diseases (MtD) represent a significant public health challenge due to their heterogenous clinical presentation, often severe and progressive symptoms, and lack of effective therapies. Environmental exposures, such bacterial and viral infection, can further compromise mitochondrial function and exacerbate the progression of MtD. Infections in MtD patients more frequently progress to sepsis, pneumonia, and other detrimental inflammatory endpoints. However, the underlying immune alterations that enhance immunopathology in MtD remain unclear, constituting a key gap in knowledge that complicates treatment and increases mortality in this vulnerable population. Here we employ in vitro and in vivo approaches to clarify the molecular and cellular basis for innate immune hyperactivity in models of polymerase gamma (Polg)-related MtD. We reveal that type I interferon (IFN-I)-mediated upregulation of caspase-11 and guanylate-binding proteins (GBPs) increase macrophage sensing of the opportunistic microbe Pseudomonas aeruginosa (PA) in Polg mutant mice. Furthermore, we show that excessive cytokine secretion and activation of pyroptotic cell death pathways contribute to lung inflammation and morbidity after infection with PA. Our work sheds new light on innate immune dysregulation in MtD and reveals potential targets for limiting infection- and inflammation-related complications in Polg-related MtD.
Collapse
Affiliation(s)
- Jordyn J. VanPortfliet
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas 77807, USA
| | - Yuanjiu Lei
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas 77807, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Camila Guerra Martinez
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas 77807, USA
| | - Jessica Wong
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | | - Kathryn Pflug
- Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, Bryan, Texas 77807, USA
| | - Raquel Sitcheran
- Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, Bryan, Texas 77807, USA
| | | | | | - Peter. J. McGuire
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Carolyn L. Cannon
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas 77807, USA
| | - A. Phillip West
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, Texas 77807, USA
| |
Collapse
|
35
|
Levee V, Sivaganesh K, Schaeffer A, Karunaratne K. POLG epilepsy presenting as new-onset refractory status epilepticus (NORSE) in pregnancy. Pract Neurol 2025; 25:56-59. [PMID: 39209381 PMCID: PMC11877079 DOI: 10.1136/pn-2024-004232] [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] [Accepted: 08/03/2024] [Indexed: 09/04/2024]
Abstract
A 21-year-old woman developed explosive new-onset refractory status epilepticus when 18 weeks pregnant. She had been previously well with no history of seizures and a normal developmental history. She had initially presented with focal impaired awareness seizures but subsequently developed status epilepticus requiring intensive care unit admission and was successfully treated with multiple anti-seizure medications. Once stabilised she was stepped down to the inpatient neurology ward and then transferred to the tertiary centre for a planned late termination of pregnancy, which was the patient's choice. Following transfer, she again developed refractory status epilepticus, requiring intensive care readmission. Subsequent investigations identified a compound heterozygous POLG genetic mutation. We discuss the challenges in the acute clinical situation and important considerations in the diagnosis and management of POLG-related epilepsy.
Collapse
Affiliation(s)
- Viva Levee
- Imperial College Healthcare NHS Trust, London, UK
| | | | - Andrew Schaeffer
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Mitochondrial Research Group, Newcastle upon Tyne, UK
| | | |
Collapse
|
36
|
Hu L, West AP, Walsh AJ. Optical metabolic imaging identifies metabolic shifts and mitochondria heterogeneity in POLG mutator macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632822. [PMID: 39868340 PMCID: PMC11760798 DOI: 10.1101/2025.01.13.632822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The polymerase gamma (POLG) gene mutation is associated with mitochondria and metabolism disorders, resulting in heterogeneous responses to immunological activation and posing challenges for mitochondrial disease therapy. Optical metabolic imaging captures the autofluorescent signal of two coenzymes, NADH and FAD, and offers a label-free approach to detect cellular metabolic phenotypes, track mitochondria morphology, and quantify metabolic heterogeneity. In this study, fluorescence lifetime imaging (FLIM) of NAD(P)H and FAD revealed that POLG mutator macrophages exhibit a decreased NAD(P)H lifetime, and optical redox ratio compared to the wild-type macrophages, indicating an increased dependence on glycolysis. FLIM revealed that both wild-type and POLG mutator macrophages switch to a decreased NAD(P)H τ 1 , and τ m after immune stimulation by Lipopolysaccharides (LPS). Furthermore, a bimodality index of subpopulation analysis identified heterogenous populations of POLG mutator macrophage responses under immune challenge by LPS. Moreover, to quantify the mitochondria variations in POLG mutator macrophages, a customized thresholding image processing pipeline was developed to segment mitochondria regions within each cell from the NADH image, allowing for the feature analysis of mitochondria clusters. Consequently, the wild-type macrophages exhibited a higher percentage of mitochondria-containing pixels and longer lengths of connected mitochondria, as compared with POLG mutated macrophages. Altogether, these results illustrate the potential of optical metabolic imaging for non-invasive detection and quantification of cellular metabolism, metabolic heterogeneity within cell populations, and intra-cellular mitochondria morphology differences in POLG mutator macrophages. Optical metabolic imaging will be valuable for studying POLG-mutation diseases and evaluating efficacy of potential therapies.
Collapse
|
37
|
Wen H, Deng H, Li B, Chen J, Zhu J, Zhang X, Yoshida S, Zhou Y. Mitochondrial diseases: from molecular mechanisms to therapeutic advances. Signal Transduct Target Ther 2025; 10:9. [PMID: 39788934 PMCID: PMC11724432 DOI: 10.1038/s41392-024-02044-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/28/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
Mitochondria are essential for cellular function and viability, serving as central hubs of metabolism and signaling. They possess various metabolic and quality control mechanisms crucial for maintaining normal cellular activities. Mitochondrial genetic disorders can arise from a wide range of mutations in either mitochondrial or nuclear DNA, which encode mitochondrial proteins or other contents. These genetic defects can lead to a breakdown of mitochondrial function and metabolism, such as the collapse of oxidative phosphorylation, one of the mitochondria's most critical functions. Mitochondrial diseases, a common group of genetic disorders, are characterized by significant phenotypic and genetic heterogeneity. Clinical symptoms can manifest in various systems and organs throughout the body, with differing degrees and forms of severity. The complexity of the relationship between mitochondria and mitochondrial diseases results in an inadequate understanding of the genotype-phenotype correlation of these diseases, historically making diagnosis and treatment challenging and often leading to unsatisfactory clinical outcomes. However, recent advancements in research and technology have significantly improved our understanding and management of these conditions. Clinical translations of mitochondria-related therapies are actively progressing. This review focuses on the physiological mechanisms of mitochondria, the pathogenesis of mitochondrial diseases, and potential diagnostic and therapeutic applications. Additionally, this review discusses future perspectives on mitochondrial genetic diseases.
Collapse
Affiliation(s)
- Haipeng Wen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Hui Deng
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xian Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
38
|
Suroviaková S, Zolak V, Igaz M, Kršiaková J, Bánovčin P. COVID-19 Infection as a Possible Trigger for POLG-Related Mitochondrial Disease: A Case Report. Cureus 2025; 17:e77592. [PMID: 39958089 PMCID: PMC11830410 DOI: 10.7759/cureus.77592] [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: 01/17/2025] [Indexed: 02/18/2025] Open
Abstract
A six-year-old child presented with an acute onset of refractory epileptic seizures during a coronavirus disease 2019 (COVID-19) infection. As her clinical condition progressed, she developed super-refractory status epilepticus, resulting in significant cognitive and motor impairments. Genetic analysis revealed a homozygous mutation in the DNA Polymerase Gamma, Catalytic Subunit (POLG) gene (c.1399G>A; p.Ala467Thr), confirming a diagnosis of Alpers-Huttenlocher syndrome. The clinical course was characterized by refractory seizures and developmental regression, and it ultimately culminated in liver failure and multiorgan dysfunction, resulting in death. This case underscores the critical importance of early genetic evaluation in children with unexplained refractory seizures, particularly for detecting underlying mitochondrial disorders such as POLG-related syndromes. Mitochondrial function is highly sensitive to physiological and environmental stressors, including viral infections. Pathogens such as hepatitis viruses, influenza virus, HIV, respiratory syncytial virus (RSV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can exacerbate mitochondrial dysfunction. Therefore, identifying genetic vulnerabilities in these patients is essential for optimizing management strategies and potentially mitigating rapid clinical decline.
Collapse
Affiliation(s)
- Stanislava Suroviaková
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Martin, SVK
| | - Vladimir Zolak
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Martin, SVK
| | - Matúš Igaz
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Martin, SVK
| | - Jana Kršiaková
- Clinic of Genetic Medicine, Jessenius Faculty of Medicine and University Hospital, Martin, SVK
| | - Peter Bánovčin
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Martin, SVK
| |
Collapse
|
39
|
Patange V, Ahirwar K, Tripathi T, Tripathi P, Shukla R. Scientific investigation of non-coding RNAs in mitochondrial epigenetic and aging disorders: Current nanoengineered approaches for their therapeutic improvement. Mitochondrion 2025; 80:101979. [PMID: 39505245 DOI: 10.1016/j.mito.2024.101979] [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: 06/25/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Genetic control is vital for the growth of cells and tissues, and it also helps living things, from single-celled organisms to complex creatures, maintain a stable internal environment. Within cells, structures called mitochondria act like tiny power plants, producing energy and keeping the cell balanced. The two primary categories of RNA are messenger RNA (mRNA) and non-coding RNA (ncRNA). mRNA carries the instructions for building proteins, while ncRNA does various jobs at the RNA level. There are different kinds of ncRNA, each with a specific role. Some help put RNA molecules together correctly, while others modify other RNAs or cut them into smaller pieces. Still others control how much protein is made from a gene. Scientists have recently discovered many more ncRNAs than previously known, and their functions are still being explored. This article analyzes the RNA molecules present within mitochondria, which have a crucial purpose in the operation of mitochondria. We'll also discuss how genes can be turned on and off without changing their DNA code, and how this process might be linked to mitochondrial RNA. Finally, we'll explore how scientists are using engineered particles to silence genes and develop new treatments based on manipulating ncRNA.
Collapse
Affiliation(s)
- Vaibhav Patange
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Kailash Ahirwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Tripti Tripathi
- Department of Physiology, Integral University, Kursi Road, Dashauli, UP 226026, India
| | - Pratima Tripathi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| |
Collapse
|
40
|
Heath O, Feichtinger RG, Achleitner MT, Hofbauer P, Mayr D, Merkevicius K, Spenger J, Steinbrücker K, Steindl C, Tiefenthaler E, Mayr JA, Wortmann SB. Mitochondrial disorder diagnosis and management- what the pediatric neurologist wants to know. Eur J Paediatr Neurol 2025; 54:75-88. [PMID: 39793294 DOI: 10.1016/j.ejpn.2024.10.009] [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: 07/23/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 01/13/2025]
Abstract
Childhood-onset mitochondrial disorders are rare genetic diseases that often manifest with neurological impairment due to altered mitochondrial structure or function. To date, pathogenic variants in 373 genes across the nuclear and mitochondrial genomes have been linked to mitochondrial disease, but the ensuing genetic and clinical complexity of these disorders poses considerable challenges to their diagnosis and management. Nevertheless, despite the current lack of curative treatment, recent advances in next generation sequencing and -omics technologies have laid the foundation for precision mitochondrial medicine through enhanced diagnostic accuracy and greater insight into pathomechanisms. This holds promise for the development of targeted treatments in this group of patients. Against a backdrop of inherent challenges and recent technological advances in mitochondrial medicine, this review discusses the current diagnostic approach to a child with suspected mitochondrial disease and outlines management considerations of particular relevance to paediatric neurologists. We highlight the importance of mitochondrial expertise centres in providing the laboratory infrastructure needed to supplement uninformative first line genomic testing with focused and/or further unbiased investigations where needed, as well as coordinating an integrated multidisciplinary model of care that is paramount to the management of patients affected by these conditions.
Collapse
Affiliation(s)
- Oliver Heath
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - René G Feichtinger
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Melanie T Achleitner
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Peter Hofbauer
- Department of Production, Landesapotheke Salzburg, Hospital Pharmacy, Salzburg, Austria
| | - Doris Mayr
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Kajus Merkevicius
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria; Clinic of Paediatrics, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania; Institute of Biosciences, Life Sciences Centre, Vilnius University, Vilnius, Lithuania
| | - Johannes Spenger
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Katja Steinbrücker
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Carina Steindl
- Institut für Klinische Psychologie der UK für Psychiatrie, Psychotherapie und Psychosomatik der PMU, Salzburg, Austria
| | - Elke Tiefenthaler
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Johannes A Mayr
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
| | - Saskia B Wortmann
- University Children's Hospital, Paracelsus Medical University (PMU), Salzburg, Austria; Amalia Children's Hospital, Department of Paediatrics, Radboudumc, Nijmegen, the Netherlands.
| |
Collapse
|
41
|
Van Haute L, Páleníková P, Tang JX, Nash PA, Simon MT, Pyle A, Oláhová M, Powell CA, Rebelo-Guiomar P, Stover A, Champion M, Deshpande C, Baple EL, Stals KL, Ellard S, Anselem O, Molac C, Petrilli G, Loeuillet L, Grotto S, Attie-Bitach T, Abdenur JE, Taylor RW, Minczuk M. Pathogenic PDE12 variants impair mitochondrial RNA processing causing neonatal mitochondrial disease. EMBO Mol Med 2025; 17:193-210. [PMID: 39567835 PMCID: PMC11729904 DOI: 10.1038/s44321-024-00172-5] [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: 04/05/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024] Open
Abstract
Pathogenic variants in either the mitochondrial or nuclear genomes are associated with a diverse group of human disorders characterized by impaired mitochondrial function. Within this group, an increasing number of families have been identified, where Mendelian genetic disorders implicate defective mitochondrial RNA biology. The PDE12 gene encodes the poly(A)-specific exoribonuclease, involved in the quality control of mitochondrial non-coding RNAs. Here, we report that disease-causing PDE12 variants in three unrelated families are associated with mitochondrial respiratory chain deficiencies and wide-ranging clinical presentations in utero and within the neonatal period, with muscle and brain involvement leading to marked cytochrome c oxidase (COX) deficiency in muscle and severe lactic acidosis. Whole exome sequencing of affected probands revealed novel, segregating bi-allelic missense PDE12 variants affecting conserved residues. Patient-derived primary fibroblasts demonstrate diminished steady-state levels of PDE12 protein, whilst mitochondrial poly(A)-tail RNA sequencing (MPAT-Seq) revealed an accumulation of spuriously polyadenylated mitochondrial RNA, consistent with perturbed function of PDE12 protein. Our data suggest that PDE12 regulates mitochondrial RNA processing and its loss results in neurological and muscular phenotypes.
Collapse
Affiliation(s)
- Lindsey Van Haute
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Petra Páleníková
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jia Xin Tang
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Pavel A Nash
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Mariella T Simon
- CHOC Children's Division of Metabolic Disorders, Orange, CA, USA
| | - Angela Pyle
- Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Monika Oláhová
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Applied Sciences, Faculty of Health & Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | | | - Pedro Rebelo-Guiomar
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Tennis Court Road, CB1 2GA, Cambridge, UK
| | - Alexander Stover
- CHOC Children's Division of Metabolic Disorders, Orange, CA, USA
| | - Michael Champion
- Department of Children's Inherited Metabolic Diseases, Evelina London Children's Hospital, Guy's & St Thomas' Hospital NHS Foundation Trust, London, UK
| | - Charulata Deshpande
- Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
- Department of Clinical Genetics, Guy's Hospital, Guy's & St Thomas' Hospital NHS Foundation Trust, London, UK
| | - Emma L Baple
- Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Karen L Stals
- Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Sian Ellard
- Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
| | - Olivia Anselem
- Maternité Port-Royal, Département de Gynécologie-Obstétrique, Hôpital Cochin Broca Hôtel-Dieu, APHP, Paris, France
| | - Clémence Molac
- Maternité Port-Royal, Département de Gynécologie-Obstétrique, Hôpital Cochin Broca Hôtel-Dieu, APHP, Paris, France
| | - Giulia Petrilli
- Service de Médecine Génomique des Maladies Rares, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Laurence Loeuillet
- Service de Médecine Génomique des Maladies Rares, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Sarah Grotto
- UF de Génétique Clinique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpital Trousseau, APHP, Paris, France
| | - Tania Attie-Bitach
- Service de Médecine Génomique des Maladies Rares, Hôpital Necker-Enfants Malades, APHP, Paris, France
- INSERM UMR 1163, Imagine Institute, Genetics and Development of the Cerebral Cortex, Université Paris Cité, Paris, France
| | - Jose E Abdenur
- CHOC Children's Division of Metabolic Disorders, Orange, CA, USA
- University of California, Irvine, Department of Pediatrics, Irvine, CA, USA
| | - Robert W Taylor
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Michal Minczuk
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| |
Collapse
|
42
|
Mabry CJ, Weindel CG, Stranahan LW, VanPortfliet JJ, Davis JR, Martinez EL, West AP, Patrick KL, Watson RO. Necrosis drives susceptibility to Mycobacterium tuberculosis in Polg D257A mutator mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603991. [PMID: 39091776 PMCID: PMC11291070 DOI: 10.1101/2024.07.17.603991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The genetic and molecular determinants that underlie the heterogeneity of Mycobacterium tuberculosis (Mtb) infection outcomes in humans are poorly understood. Multiple lines of evidence demonstrate that mitochondrial dysfunction can exacerbate mycobacterial disease severity and mutations in some mitochondrial genes confer susceptibility to mycobacterial infection in humans. Here, we report that mutations in mitochondria DNA (mtDNA) polymerase gamma (POLG) potentiate susceptibility to Mtb infection in mice. PolgD257A mutator mtDNA mice fail to mount a protective innate immune response at an early infection timepoint, evidenced by high bacterial burdens, reduced M1 macrophages, and excessive neutrophil infiltration in the lungs. Immunohistochemistry reveals signs of enhanced necrosis in the lungs of Mtb-infected PolgD257A mice and PolgD257A mutator macrophages are hyper-susceptible to extrinsic triggers of necroptosis ex vivo. By assigning a role for mtDNA mutations in driving necrosis during Mtb infection, this work further highlights the requirement for mitochondrial homeostasis in mounting balanced immune responses to Mtb.
Collapse
Affiliation(s)
- CJ Mabry
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - CG Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - LW Stranahan
- Department of Veterinary Pathobiology, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA
| | - JJ VanPortfliet
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - JR Davis
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
| | - EL Martinez
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
| | - AP West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | - KL Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - RO Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, College of Medicine, Bryan, TX 77807, USA
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
43
|
Castellotti B, Gellera C, Caputo D, Danti FR, Messina G, Corbetta M, Magri S, Taroni F, Prokisch H, Zech M, Zorzi G. Paroxysmal Non-Kinesigenic Dyskinesias Associated with Biallelic POLG Variants: A Case Report. Mov Disord 2024; 39:2300-2302. [PMID: 39404500 DOI: 10.1002/mds.30029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 12/20/2024] Open
Affiliation(s)
- Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Davide Caputo
- Unit of Developmental Neurology-Epileptology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Federica Rachele Danti
- Unit of Child Neuropsychiatry-Movement Disorder, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuliana Messina
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marinella Corbetta
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefania Magri
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Holger Prokisch
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Michael Zech
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Giovanna Zorzi
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Unit of Child Neuropsychiatry-Movement Disorder, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
44
|
Zhao Y, Zhao X, Ji K, Wang J, Zhao Y, Lin J, Gang Q, Yu M, Yuan Y, Jiang H, Sun C, Fang F, Yan C, Wang Z. The clinical and genetic spectrum of mitochondrial diseases in China: A multicenter retrospective cross-sectional study. Clin Genet 2024; 106:733-744. [PMID: 39118480 DOI: 10.1111/cge.14605] [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: 04/23/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024]
Abstract
Mitochondrial diseases (MtDs) present diverse clinical phenotypes, yet large-scale studies are hindered by their rarity. This retrospective, multicenter study, conducted across five Chinese hospitals' neurology departments from 2009 to 2019, aimed to address this gap. Nationwide, 1351 patients were enrolled, with a median onset age of 14.0 (18.5) years. The predominant phenotype was mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) (45.0%). Mitochondrial DNA (mtDNA) mutations were prevalent (87.4%), with m.3243A>G being the most common locus (48.7%). Meanwhile, POLG mutations in nuclear DNA (nDNA) accounted for 16.5%. Comparative analysis based on age groups (with a cut-off at 14 years) revealed the highest prevalence of MELAS, with Leigh syndrome (LS) and chronic progressive external ophthalmoplegia (CPEO) being the second most common phenotypes in junior and senior groups, respectively. Notably, the most commonly mutated nuclear genes varied across age groups. In conclusion, MELAS predominated in this Chinese MtD cohort, underscored by m.3243A>G and POLG as principal mtDNA mutations and pathogenic nuclear genes. The phenotypic and genotypic disparities observed among different age cohorts highlight the complex nature of MtDs.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Xutong Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Kunqian Ji
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Junling Wang
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuying Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Lin
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China
| | - Qiang Gang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Haishan Jiang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chong Sun
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China
| | - Fang Fang
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Peking University First Hospital, Beijing, China
| |
Collapse
|
45
|
Reitinger JC, Mackay DD. Optic Neuropathy Associated with POLG Mutations: A Case Series and Literature Review. J Neuroophthalmol 2024; 44:552-558. [PMID: 38294884 DOI: 10.1097/wno.0000000000002089] [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: 02/02/2024]
Abstract
BACKGROUND The clinical characteristics of patients with polymerase gamma ( POLG ) mutation-associated optic neuropathy remain incompletely characterized. METHODS We describe the clinical characteristics of 3 patients with POLG -associated optic neuropathy. We performed a literature review of optic neuropathy cases associated with POLG mutations and compared them with our cohort. RESULTS Many published cases of POLG -associated optic neuropathy in our literature review lacked details regarding severity of vision loss, visual field defects, and optical coherence tomography analysis. The clinical presentation of POLG mutations remains widely variable in age (from pediatric cases to adults) and associated systemic findings. All patients in our literature review presented with systemic symptoms, most commonly muscle weakness, ptosis, and ophthalmoplegia, whereas many young patients had severe systemic symptoms. In our case series, all 3 cases had isolated optic neuropathy affecting the papillomacular bundle, with signs such as reduced visual acuity and color vision, central visual field defects, temporal retinal nerve fiber layer loss with temporal optic disc pallor, and retinal ganglion cell complex loss. In addition, 2 of the 3 cases had added mitochondrial stressors in addition to the POLG mutation. CONCLUSIONS Clinicians should be aware that POLG mutations can present as isolated optic neuropathy primarily affecting the papillomacular bundle. With mitochondrial failure being the likely underlying pathogenic mechanism in POLG -associated optic neuropathy, helping affected patients eliminate mitochondrial stressors may be important in reducing the risk for progressive vision loss in this otherwise currently untreatable disorder.
Collapse
Affiliation(s)
- Jeremy C Reitinger
- Department of Ophthalmology (JCR), Indiana University School of Medicine, Indianapolis, Indiana; and Departments of Neurology (DDM), Ophthalmology, and Neurosurgery, Indiana University School of Medicine, Indianapolis, Indiana
| | | |
Collapse
|
46
|
Chikhaoui A, Zayoud K, Kraoua I, Bouchoucha S, Tebourbi A, Turki I, Yacoub-Youssef H. Supplementation with nicotinamide limits accelerated aging in affected individuals with cockayne syndrome and restores antioxidant defenses. Aging (Albany NY) 2024; 16:13271-13287. [PMID: 39611850 PMCID: PMC11719109 DOI: 10.18632/aging.206160] [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: 03/11/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024]
Abstract
Cockayne syndrome (CS) is a segmental progeroid syndrome characterized by defects in the DNA excision repair pathway, predisposing to neurodegenerative manifestations. It is a rare genetic disorder and an interesting model for studying premature aging. Oxidative stress and autophagy play an important role in the aging process. The study of these two processes in a model of accelerated aging and the means to counteract them would lead to the identification of relevant biomarkers with therapeutic value for healthy aging. Here we investigated the gene expression profiles of several oxidative stress-related transcripts derived from CS-affected individuals and healthy elderly donors. We also explored the effect of nicotinamide supplementation on several genes related to inflammation and autophagy. Gene expression analysis revealed alterations in two main pathways. This involves the activation of arachidonic acid metabolism and the repression of the NRF2 pathway in affected individuals with CS. The supplementation with nicotinamide adjusted these abnormalities by enhancing autophagy and decreasing inflammation. Furthermore, CSA/CSB-dependent depletion of the mitochondrial DNA polymerase-γ catalytic subunit (POLG1) was restored following nicotinamide supplementation in CS-affected individuals' fibroblasts. This study reveals the link between oxidative stress and accelerated aging in affected individuals with CS and highlights new biomarkers of cellular senescence. However, further analyses are needed to confirm these results, which could not be carried out, mainly due to the unavailability of crucial samples of this rare disease.
Collapse
Affiliation(s)
- Asma Chikhaoui
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Kouloud Zayoud
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Ichraf Kraoua
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Sami Bouchoucha
- Orthopedics Department, Béchir Hamza Children’s Hospital, Tunis 2092, Tunisia
| | - Anis Tebourbi
- Orthopedic and Trauma Surgery Department, Mongi Slim Hospital, La Marsa 2046, Tunisia
| | - Ilhem Turki
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Houda Yacoub-Youssef
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| |
Collapse
|
47
|
Karaa A, Bertini E, Carelli V, Cohen B, Ennes GM, Falk MJ, Goldstein A, Gorman G, Haas R, Hirano M, Klopstock T, Koenig MK, Kornblum C, Lamperti C, Lehman A, Longo N, Molnar MJ, Parikh S, Phan H, Pitceathly RDS, Saneto R, Scaglia F, Servidei S, Tarnopolsky M, Toscano A, Van Hove JLK, Vissing J, Vockley J, Finman JS, Abbruscato A, Brown DA, Sullivan A, Shiffer JA, Mancuso M. Genotype-specific effects of elamipretide in patients with primary mitochondrial myopathy: a post hoc analysis of the MMPOWER-3 trial. Orphanet J Rare Dis 2024; 19:431. [PMID: 39574155 PMCID: PMC11583740 DOI: 10.1186/s13023-024-03421-5] [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: 07/12/2024] [Accepted: 10/20/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND As previously published, the MMPOWER-3 clinical trial did not demonstrate a significant benefit of elamipretide treatment in a genotypically diverse population of adults with primary mitochondrial myopathy (PMM). However, the prespecified subgroup of subjects with disease-causing nuclear DNA (nDNA) pathogenic variants receiving elamipretide experienced an improvement in the six-minute walk test (6MWT), while the cohort of subjects with mitochondrial DNA (mtDNA) pathogenic variants showed no difference versus placebo. These published findings prompted additional genotype-specific post hoc analyses of the MMPOWER-3 trial. Here, we present these analyses to further investigate the findings and to seek trends and commonalities among those subjects who responded to treatment, to build a more precise Phase 3 trial design for further investigation in likely responders. RESULTS Subjects with mtDNA pathogenic variants or single large-scale mtDNA deletions represented 74% of the MMPOWER-3 population, with 70% in the mtDNA cohort having either single large-scale mtDNA deletions or MT-TL1 pathogenic variants. Most subjects in the nDNA cohort had pathogenic variants in genes required for mtDNA maintenance (mtDNA replisome), the majority of which were in POLG and TWNK. The mtDNA replisome post-hoc cohort displayed an improvement on the 6MWT, trending towards significant, in the elamipretide group when compared with placebo (25.2 ± 8.7 m versus 2.0 ± 8.6 m for placebo group; p = 0.06). The 6MWT results at week 24 in subjects with replisome variants showed a significant change in the elamipretide group subjects who had chronic progressive external ophthalmoplegia (CPEO) (37.3 ± 9.5 m versus - 8.0 ± 10.7 m for the placebo group; p = 0.0024). Pharmacokinetic (exposure-response) analyses in the nDNA cohort showed a weak positive correlation between plasma elamipretide concentration and 6MWT improvement. CONCLUSIONS Post hoc analyses indicated that elamipretide had a beneficial effect in PMM patients with mtDNA replisome disorders, underscoring the importance of considering specific genetic subtypes in PMM clinical trials. These data serve as the foundation for a follow-up Phase 3 clinical trial (NuPOWER) which has been designed as described in this paper to determine the efficacy of elamipretide in patients with mtDNA maintenance-related disorders. CLASSIFICATION OF EVIDENCE Class I CLINICALTRIALS. GOV IDENTIFIER NCT03323749.
Collapse
Affiliation(s)
- Amel Karaa
- Massachusetts General Hospital, Genetics Division Harvard Medical School Boston, Boston, MA, USA.
| | - Enrico Bertini
- Neuromuscular Unit, Bambino Gesù Ospedale Pediatrico, IRCCS, Rome, Italy
| | - Valerio Carelli
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Programma Di Neurogenetica, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Bruce Cohen
- Akron Children's Hospital, Rebecca D. Considine Research Institute, Akron, OH, USA
| | | | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Mitochondrial Medicine Frontier Program, Philadelphia, PA, USA
| | - Amy Goldstein
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Mitochondrial Medicine Frontier Program, Philadelphia, PA, USA
| | | | - Richard Haas
- University of California, San Diego, La Jolla, CA, USA
| | - Michio Hirano
- Columbia University Irving Medical Center, New York, NY, USA
| | - Thomas Klopstock
- Department of Neurology, LMU Hospital, Friedrich-Baur-Institute, Ludwig-Maximilians-Universität Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mary Kay Koenig
- Department of Pediatrics, Division of Child and Adolescent Neurology, Center for the Treatment of Pediatric Neurodegenerative Disease, University of Texas McGovern Medical School, Houston, TX, USA
| | - Cornelia Kornblum
- Department of Neurology, University Hospital of Bonn, Neuromuscular Diseases Section, Bonn, Germany
| | | | - Anna Lehman
- Vancouver General Hospital, Vancouver, BC, Canada
| | | | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Sumit Parikh
- Cleveland Clinic Neurological Institute, Cleveland, OH, USA
| | - Han Phan
- Rare Disease Research, Atlanta, GA, USA
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | | | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, TX, USA
- Joint BCM-CUHK Center of Medical Genetics, Prince of Wales Hospital, Sha Tin, Hong Kong SAR, China
| | - Serenella Servidei
- Fondazione Policlinico Universitario A. Gemelli and Istituto Di Neurologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mark Tarnopolsky
- Division of Neuromuscular and Neurometabolic Disorders, McMaster University Children's Hospital, Hamilton, ON, Canada
| | - Antonio Toscano
- Department of Clinical and Experimental Medicine, ERN-NMD Center for Neuromuscular Disorders of Messina, University of Messina, Messina, Italy
| | - Johan L K Van Hove
- University of Colorado and Children's Hospital Colorado, Aurora, CO, USA
| | - John Vissing
- Copenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jerry Vockley
- Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | | - Michelango Mancuso
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Pisa, Italy
| |
Collapse
|
48
|
Hu W, Shi C, Guo H, Zhang B. POLG p.A962T Mutation Leads to Neuronal Mitochondrial Dysfunction That is Restored After Mitochondrial Transplantation. Physiol Res 2024; 73:801-808. [PMID: 39545794 PMCID: PMC11629961 DOI: 10.33549/physiolres.935313] [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: 01/02/2024] [Accepted: 05/14/2024] [Indexed: 12/13/2024] Open
Abstract
Mutations in DNA polymerase gamma (POLG) are known as the predominant cause of inherited mitochondrial disorders. But how these POLG mutations disturb mitochondrial function remains to be determined. Furthermore, no effective therapy, to date, has been reported for POLG diseases. Using differentiated SH-SY5Y cells, a human neuronal model cell line, the current study investigated whether the novel POLG variant p.A962T impairs mitochondrial function. This involved quantifying mitochondrial DNA (mtDNA) content using PCR and assessing the expression levels of the subunits of complex IV (COXI-IV), a complex I subunit NDUFV1 and Cytochrome C (Cyto C) release using Western blotting. Activities of mitochondrial complex I, II, and IV were measured using colorimetric assays. Mitochondrial membrane potential (delta Psim) and ATP were evaluated using fluorescence assays and luminescent assays, respectively. In addition, we investigated whether mitochondrial transplantation (MT) using Pep-1-conjugated mitochondria could compensate for mitochondrial defects caused by the variant in cells carrying mutant POLG. The results of this study showed that POLG p.A962T mutation resulted in mitochondrial defects, including mitochondrial DNA (mtDNA) depletion, membrane potential (delta Psim) depolarization and adenosine triphosphate (ATP) reduction. Mechanistically, POLG mutation-caused mtDNA depletion led to the loss of mtDNA-encoded subunits of complex I and IV and thus compromised their activities. POLG p.A962T mutation is a pathogenic mutation leading to mitochondrial malfunction and mtDNA depletion in neurons. Cell-penetrating peptide Pep-1-mediated MT treatment compensated for mitochondrial defects induced by these POLG variants, suggesting the therapeutic application of this method in POLG diseases.
Collapse
Affiliation(s)
- W Hu
- Department of Pharmacy, Yiyang Medical College, Yiyang, China; College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA.
| | | | | | | |
Collapse
|
49
|
Liu Z, Xie Y, Lou X, Zeng X, Zhang L, Yu M, Wang J, Li J, Shen D, Li H, Zhao S, Zhou Y, Fang H, Lyu J, Yuan Y, Wang Z, Jin L, Fang F. A novel m.5906G > a variant in MT-CO1 causes MELAS/Leigh overlap syndrome. Mol Genet Genomics 2024; 299:102. [PMID: 39460813 DOI: 10.1007/s00438-024-02181-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 09/02/2024] [Indexed: 10/28/2024]
Abstract
The MELAS/Leigh overlap syndrome manifests with a blend of clinical and radiographic traits from both MELAS and LS. However, the association of MELAS/Leigh overlap syndrome with MT-CO1 gene variants has not been previously reported. In this study, we report a patient diagnosed with MELAS/Leigh overlap syndrome harboring the m.5906G > A variant in MT-CO1, with biochemical evidence supporting the pathogenicity of the variant. The variant m.5906G > A that led to a synonymous variant in the start codon of MT-CO1 was filtered as the candidate disease-causing variant of the patient. Patient-derived fibroblasts were used to generate a series of monoclonal cells carrying different m.5906G > A variant loads for further functional assays. The oxygen consumption rate, ATP production, mitochondrial membrane potential and lactate assay indicated an impairment of cellular bioenergetics due to the m.5906G > A variant. Blue native PAGE analysis revealed that the m.5906G > A variant caused a deficiency in the content of mitochondrial oxidative phosphorylation complexes. Furthermore, molecular biology assays performed for the pathogenesis, mtDNA copy number, mtDNA-encoded subunits, and recovery capacity of mtDNA were all deficient due to the m.5906G > A variant, which might be caused by mtDNA replication deficiency. Overall, our findings demonstrated the pathogenicity of m.5906G > A variant and proposed a potential pathogenic mechanism, thereby expanding the genetic spectrum of MELAS/Leigh overlap syndrome.
Collapse
Affiliation(s)
- Zhimei Liu
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Yaojun Xie
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiaoting Lou
- Laboratory Medicine Center, Department of Genetic and Genomic Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xiaofei Zeng
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Luyi Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Junling Wang
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jiuwei Li
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Danmin Shen
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Hua Li
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Suzhou Zhao
- Fujungenetics Technologies Co, Ltd, Beijing, 100176, China
| | - Yuwei Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hezhi Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jianxin Lyu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China.
| | - Liqin Jin
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Department of Scientific Research, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Fang Fang
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
50
|
Sun H, Zhang G, Li N, Bu X. Molecular diagnosis of patients with syndromic short stature identified by trio whole-exome sequencing. Front Genet 2024; 15:1399186. [PMID: 39415983 PMCID: PMC11479978 DOI: 10.3389/fgene.2024.1399186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background Short stature is a complex disorder with phenotypic and genetic heterogeneity. This study aimed to investigate clinical phenotypes and molecular basis of a cohort of patients with short stature. Methods Trio whole-exome sequencing (Trio-WES) was performed to explore the genetic aetiology and obtain a molecular diagnosis in twenty Chinese probands with syndromic and isolated short stature. Results Of the twenty probands, six (6/20, 30%) patients with syndromic short stature obtained a molecular diagnosis. One novel COMP pathogenic variant c.1359delC, p.N453fs*62 and one LZTR1 likely pathogenic variant c.509G>A, p.R170Q were identified in a patient with short stature and skeletal dysplasia. One novel de novo NAA15 pathogenic variant c.63T>G, p.Y21X and one novel de novo KMT2A pathogenic variant c.3516T>A, p.N1172K was identified in two probands with short stature, intellectual disability and abnormal behaviours, respectively. One patient with short stature, cataract, and muscle weakness had a de novo POLG pathogenic variant c.2863 T>C, p.Y955H. One PHEX pathogenic variant c.1104G>A, p.W368X was identified in a patient with short stature and rickets. Maternal uniparental disomy 7 (mUPD7) was pathogenic in a patient with pre and postnatal growth retardation, wide forehead, triangular face, micrognathia and clinodactyly. Thirteen patients with isolated short stature had negative results. Conclusion Trio-WES is an important strategy for identifying genetic variants and UPD in patients with syndromic short stature, in which dual genetic variants are existent in some individuals. It is important to differentiate between syndromic and isolated short stature. Genetic testing has a high yield for syndromic patients but low for isolated patients.
Collapse
Affiliation(s)
- Huihui Sun
- Department of Paediatrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Geng Zhang
- Beijing Chigene Translational Medical Research Center Company, Beijing, China
| | - Na Li
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Xiangfang Bu
- Department of Paediatrics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|