1
|
Xu IR, Danzi MC, Raposo J, Züchner S. The continued promise of genomic technologies and software in neurogenetics. J Neuromuscul Dis 2025:22143602251325345. [PMID: 40208247 DOI: 10.1177/22143602251325345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
The continued evolution of genomic technologies over the past few decades has revolutionized the field of neurogenetics, offering profound insights into the genetic underpinnings of neurological disorders. Identification of causal genes for numerous monogenic neurological conditions has informed key aspects of disease mechanisms and facilitated research into critical proteins and molecular pathways, laying the groundwork for therapeutic interventions. However, the question remains: has this transformative trend reached its zenith? In this review, we suggest that despite significant strides in genome sequencing and advanced computational analyses, there is still ample room for methodological refinement. We anticipate further major genetic breakthroughs corresponding with the increased use of long-read genomes, variant calling software, AI tools, and data aggregation databases. Genetic progress has historically been driven by technological advancements from the commercial sector, which are developed in response to academic research needs, creating a continuous cycle of innovation and discovery. This review explores the potential of genomic technologies to address the challenges of neurogenetic disorders. By outlining both established and modern resources, we aim to emphasize the importance of genetic technologies as we enter an era poised for discoveries.
Collapse
Affiliation(s)
- Isaac Rl Xu
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matt C Danzi
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jacquelyn Raposo
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
2
|
Strupp W. A new variant of the electromagnetic field theory of consciousness: approaches to empirical confirmation. Front Neurol 2024; 15:1420676. [PMID: 39494171 PMCID: PMC11527664 DOI: 10.3389/fneur.2024.1420676] [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: 06/04/2024] [Accepted: 09/16/2024] [Indexed: 11/05/2024] Open
Abstract
There are various electromagnetic (EM) field theories of consciousness. They postulate an epineural EM field which, due to its binding properties, unifies the different neuronal information differences originating from various sensory and cognitive processes. Only through a real physical integration in space within this field could phenomenal consciousness arise. This would solve the binding problem mentioned in the philosophy of mind. On closer inspection, the electromagnetic interaction not only provides an explanation for the integrative property of the EM field, but also for the necessary differentiating contrasts of information. This article will take a closer look at the physical properties of a postulated EM field. It will also show how the problem of qualia in connection with emergentism could be solved by a new variant of EM field theory. If it can be clearly demonstrated that the postulated epineural EM field plays a decisive role in the origin of consciousness in addition to neuronal "wired" information processing, this also leaves less room for metaphysical assumptions that attempt to solve the binding problem. In experiments to prove the postulated epineural EM field by means of external electromagnetic manipulations, it can never be ruled out that these also have a direct effect on the "wired" neuronal signal processing. Therefore, on the way to proving the EM field theory of consciousness, an experimental method is needed that must ensure that external manipulations only affect the extensions of the EM field without directly influencing the neuronal network. A method will be discussed here that works with the shielding of EM fields instead of external electromagnetic stimuli.
Collapse
|
3
|
Mavillard F, Guerra-Castellano A, Guerrero-Gómez D, Rivas E, Cantero G, Servian-Morilla E, Folland C, Ravenscroft G, Martín MA, Miranda-Vizuete A, Cabrera-Serrano M, Diaz-Moreno I, Paradas C. A splice-altering homozygous variant in COX18 causes severe sensory-motor neuropathy with oculofacial apraxia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167330. [PMID: 38960055 DOI: 10.1016/j.bbadis.2024.167330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Affiliation(s)
- Fabiola Mavillard
- Neuromuscular Unit, Neurology Department, Instituto de Biomedicina de Sevilla (IBIS)/Hospital Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Sevilla, Spain
| | | | - David Guerrero-Gómez
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla (IBIS)/Hospital Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Eloy Rivas
- Department of Neuropathology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Gloria Cantero
- Neuromuscular Unit, Neurology Department, Instituto de Biomedicina de Sevilla (IBIS)/Hospital Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Sevilla, Spain
| | - Emilia Servian-Morilla
- Neuromuscular Unit, Neurology Department, Instituto de Biomedicina de Sevilla (IBIS)/Hospital Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Sevilla, Spain
| | - Chiara Folland
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, WA 6009, Australia
| | - Gianina Ravenscroft
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, WA 6009, Australia
| | - Miguel A Martín
- Mitochondrial & Neuromuscular Disorders Group, Genetics Department, Hospital 12 de Octubre Research Institute (imas12), Madrid, Spain; Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Miranda-Vizuete
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla (IBIS)/Hospital Virgen del Rocío/CSIC/Universidad de Sevilla, Spain
| | - Macarena Cabrera-Serrano
- Neuromuscular Unit, Neurology Department, Instituto de Biomedicina de Sevilla (IBIS)/Hospital Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Sevilla, Spain
| | - Irene Diaz-Moreno
- Instituto de Investigaciones Químicas, Universidad de Sevilla-CSIC, Sevilla, Spain.
| | - Carmen Paradas
- Neuromuscular Unit, Neurology Department, Instituto de Biomedicina de Sevilla (IBIS)/Hospital Virgen del Rocío/CSIC/Universidad de Sevilla, Spain; Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Sevilla, Spain.
| |
Collapse
|
4
|
Armirola-Ricaurte C, Morant L, Adant I, Hamed SA, Pipis M, Efthymiou S, Amor-Barris S, Atkinson D, Van de Vondel L, Tomic A, de Vriendt E, Zuchner S, Ghesquiere B, Hanna M, Houlden H, Lunn MP, Reilly MM, Rasic VM, Jordanova A. Biallelic variants in COX18 cause a mitochondrial disorder primarily manifesting as peripheral neuropathy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.03.24309787. [PMID: 39006432 PMCID: PMC11245062 DOI: 10.1101/2024.07.03.24309787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Defects in mitochondrial dynamics are a common cause of Charcot-Marie-Tooth disease (CMT), while primary deficiencies in the mitochondrial respiratory chain (MRC) are rare and atypical for this etiology. This study aims to report COX18 as a novel CMT-causing gene. This gene encodes an assembly factor of mitochondrial Complex IV (CIV) that translocates the C-terminal tail of MTCO2 across the mitochondrial inner membrane. Exome sequencing was performed in four affected individuals. The patients and available family members underwent thorough neurological and electrophysiological assessment. The impact of one of the identified variants on splicing, protein levels, and mitochondrial bioenergetics was investigated in patient-derived lymphoblasts. The functionality of the mutant protein was assessed using a Proteinase K protection assay and immunoblotting. Neuronal relevance of COX18 was assessed in a Drosophila melanogaster knockdown model. Exome sequencing coupled with homozygosity mapping revealed a homozygous splice variant c.435-6A>G in COX18 in two siblings with early-onset progressive axonal sensory-motor peripheral neuropathy. By querying external databases, we identified two additional families with rare deleterious biallelic variants in COX18 . All affected individuals presented with axonal CMT and some patients also exhibited central nervous system symptoms, such as dystonia and spasticity. Functional characterization of the c.435-6A>G variant demonstrated that it leads to the expression of an alternative transcript that lacks exon 2, resulting in a stable but defective COX18 isoform. The mutant protein impairs CIV assembly and activity, leading to a reduction in mitochondrial membrane potential. Downregulation of the COX18 homolog in Drosophila melanogaster displayed signs of neurodegeneration, including locomotor deficit and progressive axonal degeneration of sensory neurons. Our study presents genetic and functional evidence that supports COX18 as a newly identified gene candidate for autosomal recessive axonal CMT with or without central nervous system involvement. These findings emphasize the significance of peripheral neuropathy within the spectrum of primary mitochondrial disorders and the role of mitochondrial CIV in the development of CMT. Our research has important implications for the diagnostic workup of CMT patients.
Collapse
|
5
|
Armirola-Ricaurte C, Zonnekein N, Koutsis G, Amor-Barris S, Pelayo-Negro AL, Atkinson D, Efthymiou S, Turchetti V, Dinopoulos A, Garcia A, Karakaya M, Moris G, Polat AI, Yiş U, Espinos C, Van de Vondel L, De Vriendt E, Karadima G, Wirth B, Hanna M, Houlden H, Berciano J, Jordanova A. Alternative splicing expands the clinical spectrum of NDUFS6-related mitochondrial disorders. Genet Med 2024; 26:101117. [PMID: 38459834 PMCID: PMC11180951 DOI: 10.1016/j.gim.2024.101117] [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: 10/17/2023] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024] Open
Abstract
PURPOSE We describe 3 families with Charcot-Marie-Tooth neuropathy (CMT), harboring a homozygous NDUFS6 NM_004553.6:c.309+5G>A variant previously linked to fatal Leigh syndrome. We aimed to characterize clinically and molecularly the newly identified patients and understand the mechanism underlying their milder phenotype. METHODS The patients underwent extensive clinical examinations. Exome sequencing was done in 4 affected individuals. The functional effect of the c.309+5G>A variant was investigated in patient-derived EBV-transformed lymphoblasts at the complementary DNA, protein, and mitochondrial level. Alternative splicing was evaluated using complementary DNA long-read sequencing. RESULTS All patients presented with early-onset, slowly progressive axonal CMT, and nystagmus; some exhibited additional central nervous system symptoms. The c.309+5G>A substitution caused the expression of aberrantly spliced transcripts and negligible levels of the canonical transcript. Immunoblotting showed reduced levels of mutant isoforms. No detectable defects in mitochondrial complex stability or bioenergetics were found. CONCLUSION We expand the clinical spectrum of NDUFS6-related mitochondrial disorders to include axonal CMT, emphasizing the clinical and pathophysiologic overlap between these 2 clinical entities. This work demonstrates the critical role that alternative splicing may play in modulating the severity of a genetic disorder, emphasizing the need for careful consideration when interpreting splice variants and their implications on disease prognosis.
Collapse
Affiliation(s)
- Camila Armirola-Ricaurte
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Noortje Zonnekein
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Georgios Koutsis
- Neurogenetics Unit, 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Silvia Amor-Barris
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Ana Lara Pelayo-Negro
- University Hospital Marqués de Valdecilla (IFIMAV), University of Cantabria, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Santander, Spain
| | - Derek Atkinson
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Valentina Turchetti
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Argyris Dinopoulos
- 3rd Department of Pediatrics, Attiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonio Garcia
- Service of Clinical Neurophysiology, University Hospital Marqués de Valdecilla, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Santander, Spain
| | - Mert Karakaya
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - German Moris
- Service of Neurology, University Hospital Central de Asturias, University of Oviedo, Oviedo, Spain
| | - Ayşe Ipek Polat
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Uluç Yiş
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Carmen Espinos
- Rare Neurodegenerative Disease Laboratory, Centro de Investigación Príncipe Felipe (CIPF), CIBER on Rare Diseases (CIBERER), Valencia, Spain
| | - Liedewei Van de Vondel
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Els De Vriendt
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Georgia Karadima
- Neurogenetics Unit, 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Michael Hanna
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Jose Berciano
- University Hospital Marqués de Valdecilla (IFIMAV), University of Cantabria, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Santander, Spain
| | - Albena Jordanova
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Department of Medical Chemistry and Biochemistry, Medical University-Sofia, Sofia, Bulgaria.
| |
Collapse
|
6
|
Rebelo AP, Tomaselli PJ, Medina J, Wang Y, Dohrn MF, Nyvltova E, Danzi MC, Garrett M, Smith SE, Pestronk A, Li C, Ruiz A, Jacobs E, Feely SME, França MC, Gomes MV, Santos DF, Kumar S, Lombard DB, Saporta M, Hekimi S, Barrientos A, Weihl C, Shy ME, Marques W, Zuchner S. Biallelic variants in COQ7 cause distal hereditary motor neuropathy with upper motor neuron signs. Brain 2023; 146:4191-4199. [PMID: 37170631 PMCID: PMC10545612 DOI: 10.1093/brain/awad158] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 04/12/2023] [Accepted: 04/23/2023] [Indexed: 05/13/2023] Open
Abstract
COQ7 encodes a hydroxylase responsible for the penultimate step of coenzyme Q10 (CoQ10) biosynthesis in mitochondria. CoQ10 is essential for multiple cellular functions, including mitochondrial oxidative phosphorylation, lipid metabolism, and reactive oxygen species homeostasis. Mutations in COQ7 have been previously associated with primary CoQ10 deficiency, a clinically heterogeneous multisystemic mitochondrial disorder. We identified COQ7 biallelic variants in nine families diagnosed with distal hereditary motor neuropathy with upper neuron involvement, expending the clinical phenotype associated with defects in this gene. A recurrent p.Met1? change was identified in five families from Brazil with evidence of a founder effect. Fibroblasts isolated from patients revealed a substantial depletion of COQ7 protein levels, indicating protein instability leading to loss of enzyme function. High-performance liquid chromatography assay showed that fibroblasts from patients had reduced levels of CoQ10, and abnormal accumulation of the biosynthetic precursor DMQ10. Accordingly, fibroblasts from patients displayed significantly decreased oxygen consumption rates in patients, suggesting mitochondrial respiration deficiency. Induced pluripotent stem cell-derived motor neurons from patient fibroblasts showed significantly increased levels of extracellular neurofilament light protein, indicating axonal degeneration. Our findings indicate a molecular pathway involving CoQ10 biosynthesis deficiency and mitochondrial dysfunction in patients with distal hereditary motor neuropathy. Further studies will be important to evaluate the potential benefits of CoQ10 supplementation in the clinical outcome of the disease.
Collapse
Affiliation(s)
- Adriana P Rebelo
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Pedro J Tomaselli
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Jessica Medina
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Ying Wang
- Department of Biology, McGill University, Montreal, QC, H3A 1A1, Canada
| | - Maike F Dohrn
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen 52074, Germany
| | - Eva Nyvltova
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matt C Danzi
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Mark Garrett
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Sean E Smith
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Alan Pestronk
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Chengcheng Li
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Ariel Ruiz
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Elizabeth Jacobs
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Shawna M E Feely
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Marcondes C França
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Marcus V Gomes
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Diogo F Santos
- Department of Neurology, Federal University of Uberlândia, Uberlândia, MG 38405-320, Brazil
| | - Surinder Kumar
- Department of Pathology & Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David B Lombard
- Department of Pathology & Laboratory Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mario Saporta
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Siegfried Hekimi
- Department of Biology, McGill University, Montreal, QC, H3A 1A1, Canada
| | - Antoni Barrientos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Conrad Weihl
- Department of Neurology, Washington University, St. Louis, MO 63112, USA
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Wilson Marques
- Department of Neurology, University of São Paulo, Ribeirão Preto, 14048-900, Brazil
| | - Stephan Zuchner
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
7
|
Hsueh HW, Kao HJ, Chao CC, Hsueh SJ, Huang YN, Lin WJ, Su JP, Shy HT, Yeh TY, Lin CC, Kwok PY, Lee NC, Hsieh ST. Identification of an 85-kb Heterozygous 4p Microdeletion With Full Genome Analysis in Autosomal Dominant Charcot-Marie-Tooth Disease. Neurol Genet 2023; 9:e200078. [PMID: 37346931 PMCID: PMC10281236 DOI: 10.1212/nxg.0000000000200078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/06/2023] [Indexed: 06/23/2023]
Abstract
Background and Objectives Charcot-Marie-Tooth disease (CMT) is a syndrome of a hereditary neurodegenerative condition affecting the peripheral nervous system and is a single gene disorder. Deep phenotyping coupled with advanced genetic techniques is critical in discovering new genetic defects of rare genetic disorders such as CMT. Methods We applied multidisciplinary investigations to examine the neurophysiology and nerve pathology in a family that fulfilled the diagnosis of CMT2. When phenotype-guided first-tier genetic tests and whole-exome sequencing did not yield a molecular diagnosis, we conducted full genome analysis by examining phased whole-genome sequencing and whole-genome optical mapping data to search for the causal variation. We then performed a systematic review to compare the reported patients with interstitial microdeletion in the short arm of chromosome 4. Results In this family with CMT2, we reported the discovery of a heterozygous 85-kb microdeletion in the short arm of chromosome 4 (4p16.3)[NC_000004.12:g.1733926_1819031del] spanning 3 genes [TACC3 (intron 6-exon 16), FGFR3 (total deletion), and LETM1 (intron 10-exon14)] that cosegregated with disease phenotypes in family members. The clinical features of peripheral nerve degeneration in our family are distinct from the well-known 4p microdeletion syndrome of Wolf-Hirschhorn syndrome, in which brain involvement is the major phenotype. Discussion In summary, we used the full genome analysis approach to discover a new microdeletion in a family with CMT2. The deleted segment contains 3 genes (TACC3, FGFR3, and LETM1) that likely play a role in the pathogenesis of nerve degeneration.
Collapse
Affiliation(s)
- Hsueh Wen Hsueh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Hsiao-Jung Kao
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Chi-Chao Chao
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Sung-Ju Hsueh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Yu-Ning Huang
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Wan-Jia Lin
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Jen-Ping Su
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Horng-Tzer Shy
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Ti-Yen Yeh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Cheng-Chen Lin
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Pui-Yan Kwok
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Ni-Chung Lee
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Sung-Tsang Hsieh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| |
Collapse
|
8
|
Wei J, Wang S, Zhu H, Cui W, Gao J, Gao C, Yu B, Liu B, Chen J, Peng J. Hepatic depletion of nucleolar protein mDEF causes excessive mitochondrial copper accumulation associated with p53 and NRF1 activation. iScience 2023; 26:107220. [PMID: 37456842 PMCID: PMC10339200 DOI: 10.1016/j.isci.2023.107220] [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: 03/03/2023] [Revised: 05/15/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Copper is an essential component in the mitochondrial respiratory chain complex IV (cytochrome c oxidases). However, whether any nucleolar factor(s) is(are) involved in regulating the mitochondrial copper homeostasis remains unclear. The nucleolar localized Def-Capn3 protein degradation pathway cleaves target proteins, including p53, in both zebrafish and human nucleoli. Here, we report that hepatic depletion of mDEF in mice causes an excessive copper accumulation in the mitochondria. We find that mDEF-depleted hepatocytes show an exclusion of CAPN3 from the nucleoli and accumulate p53 and NRF1 proteins in the nucleoli. Furthermore, we find that NRF1 is a CAPN3 substrate. Elevated p53 and NRF1 enhances the expression of Sco2 and Cox genes, respectively, to allow more copper acquirement in the mDefloxp/loxp, Alb:Cre mitochondria. Our findings reveal that the mDEF-CAPN3 pathway serves as a novel mechanism for regulating the mitochondrial copper homeostasis through targeting its substrates p53 and NRF1.
Collapse
Affiliation(s)
- Jinsong Wei
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shuai Wang
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Haozhe Zhu
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Cui
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianan Gao
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ce Gao
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bojing Liu
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jun Chen
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jinrong Peng
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
9
|
Ma Z, Liang H, Cui R, Ji J, Liu H, Liu X, Shen P, Wang H, Wang X, Song Z, Jiang Y. Construction of a risk model and prediction of prognosis and immunotherapy based on cuproptosis-related LncRNAs in the urinary system pan-cancer. Eur J Med Res 2023; 28:198. [PMID: 37370148 DOI: 10.1186/s40001-023-01173-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Urinary pan-cancer system is a general term for tumors of the urinary system including renal cell carcinoma (RCC), prostate cancer (PRAD), and bladder cancer (BLCA). Their location, physiological functions, and metabolism are closely related, making the occurrence and outcome of these tumors highly similar. Cuproptosis is a new type of cell death that is different from apoptosis and plays an essential role in tumors. Therefore, it is necessary to study the molecular mechanism of cuproptosis-related lncRNAs to urinary system pan-cancer for the prognosis, clinical diagnosis, and treatment of urinary tumors. METHOD In our study, we identified 35 co-expression cuproptosis-related lncRNAs (CRLs) from the urinary pan-cancer system. 28 CRLs were identified as prognostic-related CRLs by univariate Cox regression analysis. Then 12 CRLs were obtained using lasso regression and multivariate cox analysis to construct a prognostic model. We divided patients into high- and low-risk groups based on the median risk scores. Next, Kaplan-Meier analysis, principal component analysis (PCA), functional rich annotations, and nomogram were used to compare the differences between the high- and low-risk groups. Finally, the prediction of tumor immune dysfunction and rejection, gene mutation, and drug sensitivity were discussed. CONCLUSION Finally, the candidate molecules of the urinary system pan-cancer were identified. This CRLs risk model may be promising for clinical prediction of prognosis and immunotherapy response in urinary system pan-cancer patients.
Collapse
Affiliation(s)
- Zhihui Ma
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Haining Liang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Rongjun Cui
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Jinli Ji
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Hongfeng Liu
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xiaoxue Liu
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Ping Shen
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Huan Wang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Xingyun Wang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Zheyao Song
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Ying Jiang
- Mudanjiang Medical University, Mudanjiang, Heilongjiang, China.
| |
Collapse
|
10
|
Zhang YY, Li XS, Ren KD, Peng J, Luo XJ. Restoration of metal homeostasis: a potential strategy against neurodegenerative diseases. Ageing Res Rev 2023; 87:101931. [PMID: 37031723 DOI: 10.1016/j.arr.2023.101931] [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: 01/31/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Metal homeostasis is critical to normal neurophysiological activity. Metal ions are involved in the development, metabolism, redox and neurotransmitter transmission of the central nervous system (CNS). Thus, disturbance of homeostasis (such as metal deficiency or excess) can result in serious consequences, including neurooxidative stress, excitotoxicity, neuroinflammation, and nerve cell death. The uptake, transport and metabolism of metal ions are highly regulated by ion channels. There is growing evidence that metal ion disorders and/or the dysfunction of ion channels contribute to the progression of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Therefore, metal homeostasis-related signaling pathways are emerging as promising therapeutic targets for diverse neurological diseases. This review summarizes recent advances in the studies regarding the physiological and pathophysiological functions of metal ions and their channels, as well as their role in neurodegenerative diseases. In addition, currently available metal ion modulators and in vivo quantitative metal ion imaging methods are also discussed. Current work provides certain recommendations based on literatures and in-depth reflections to improve neurodegenerative diseases. Future studies should turn to crosstalk and interactions between different metal ions and their channels. Concomitant pharmacological interventions for two or more metal signaling pathways may offer clinical advantages in treating the neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xi-Sheng Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013,China
| | - Kai-Di Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013,China.
| |
Collapse
|
11
|
Higuchi Y, Takashima H. Clinical genetics of Charcot-Marie-Tooth disease. J Hum Genet 2023; 68:199-214. [PMID: 35304567 DOI: 10.1038/s10038-022-01031-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/02/2022] [Accepted: 03/06/2022] [Indexed: 02/08/2023]
Abstract
Recent research in the field of inherited peripheral neuropathies (IPNs) such as Charcot-Marie-Tooth (CMT) disease has helped identify the causative genes provided better understanding of the pathogenesis, and unraveled potential novel therapeutic targets. Several reports have described the epidemiology, clinical characteristics, molecular pathogenesis, and novel causative genes for CMT/IPNs in Japan. Based on the functions of the causative genes identified so far, the following molecular and cellular mechanisms are believed to be involved in the causation of CMTs/IPNs: myelin assembly, cytoskeletal structure, myelin-specific transcription factor, nuclear related, endosomal sorting and cell signaling, proteasome and protein aggregation, mitochondria-related, motor proteins and axonal transport, tRNA synthetases and RNA metabolism, and ion channel-related mechanisms. In this article, we review the epidemiology, genetic diagnosis, and clinicogenetic characteristics of CMT in Japan. In addition, we discuss the newly identified novel causative genes for CMT/IPNs in Japan, namely MME and COA7. Identification of the new causes of CMT will facilitate in-depth characterization of the underlying molecular mechanisms of CMT, leading to the establishment of therapeutic approaches such as drug development and gene therapy.
Collapse
Affiliation(s)
- Yujiro Higuchi
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiroshi Takashima
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.
| |
Collapse
|
12
|
Horvath R, Medina J, Reilly MM, Shy ME, Zuchner S. Peripheral neuropathy in mitochondrial disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 194:99-116. [PMID: 36813324 DOI: 10.1016/b978-0-12-821751-1.00014-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Mitochondria are essential for the health and viability of both motor and sensory neurons and their axons. Processes that disrupt their normal distribution and transport along axons will likely cause peripheral neuropathies. Similarly, mutations in mtDNA or nuclear encoded genes result in neuropathies that either stand alone or are part of multisystem disorders. This chapter focuses on the more common genetic forms and characteristic clinical phenotypes of "mitochondrial" peripheral neuropathies. We also explain how these various mitochondrial abnormalities cause peripheral neuropathy. In a patient with a neuropathy either due to a mutation in a nuclear or an mtDNA gene, clinical investigations aim to characterize the neuropathy and make an accurate diagnosis. In some patients, this may be relatively straightforward, where a clinical assessment and nerve conduction studies followed by genetic testing is all that is needed. In others, multiple investigations including a muscle biopsy, CNS imaging, CSF analysis, and a wide range of metabolic and genetic tests in blood and muscle may be needed to establish diagnosis.
Collapse
Affiliation(s)
- Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, John van Geest Centre for Brain Repair, Cambridge, United Kingdom.
| | - Jessica Medina
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mary M Reilly
- MRC Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Stephan Zuchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
13
|
Protein Transduction Domain-Mediated Delivery of Recombinant Proteins and In Vitro Transcribed mRNAs for Protein Replacement Therapy of Human Severe Genetic Mitochondrial Disorders: The Case of Sco2 Deficiency. Pharmaceutics 2023; 15:pharmaceutics15010286. [PMID: 36678915 PMCID: PMC9861957 DOI: 10.3390/pharmaceutics15010286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Mitochondrial disorders represent a heterogeneous group of genetic disorders with variations in severity and clinical outcomes, mostly characterized by respiratory chain dysfunction and abnormal mitochondrial function. More specifically, mutations in the human SCO2 gene, encoding the mitochondrial inner membrane Sco2 cytochrome c oxidase (COX) assembly protein, have been implicated in the mitochondrial disorder fatal infantile cardioencephalomyopathy with COX deficiency. Since an effective treatment is still missing, a protein replacement therapy (PRT) was explored using protein transduction domain (PTD) technology. Therefore, the human recombinant full-length mitochondrial protein Sco2, fused to TAT peptide (a common PTD), was produced (fusion Sco2 protein) and successfully transduced into fibroblasts derived from a SCO2/COX-deficient patient. This PRT contributed to effective COX assembly and partial recovery of COX activity. In mice, radiolabeled fusion Sco2 protein was biodistributed in the peripheral tissues of mice and successfully delivered into their mitochondria. Complementary to that, an mRNA-based therapeutic approach has been more recently considered as an innovative treatment option. In particular, a patented, novel PTD-mediated IVT-mRNA delivery platform was developed and applied in recent research efforts. PTD-IVT-mRNA of full-length SCO2 was successfully transduced into the fibroblasts derived from a SCO2/COX-deficient patient, translated in host ribosomes into a nascent chain of human Sco2, imported into mitochondria, and processed to the mature protein. Consequently, the recovery of reduced COX activity was achieved, thus suggesting the potential of this mRNA-based technology for clinical translation as a PRT for metabolic/genetic disorders. In this review, such research efforts will be comprehensibly presented and discussed to elaborate their potential in clinical application and therapeutic usefulness.
Collapse
|
14
|
Garza NM, Swaminathan AB, Maremanda KP, Zulkifli M, Gohil VM. Mitochondrial copper in human genetic disorders. Trends Endocrinol Metab 2023; 34:21-33. [PMID: 36435678 PMCID: PMC9780195 DOI: 10.1016/j.tem.2022.11.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022]
Abstract
Copper is an essential micronutrient that serves as a cofactor for enzymes involved in diverse physiological processes, including mitochondrial energy generation. Copper enters cells through a dedicated copper transporter and is distributed to intracellular cuproenzymes by copper chaperones. Mitochondria are critical copper-utilizing organelles that harbor an essential cuproenzyme cytochrome c oxidase, which powers energy production. Mutations in copper transporters and chaperones that perturb mitochondrial copper homeostasis result in fatal genetic disorders. Recent studies have uncovered the therapeutic potential of elesclomol, a copper ionophore, for the treatment of copper deficiency disorders such as Menkes disease. Here we review the role of copper in mitochondrial energy metabolism in the context of human diseases and highlight the recent developments in copper therapeutics.
Collapse
Affiliation(s)
- Natalie M Garza
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Abhinav B Swaminathan
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Krishna P Maremanda
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Mohammad Zulkifli
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Vishal M Gohil
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
15
|
Wang L, Yang Z, He X, Pu S, Yang C, Wu Q, Zhou Z, Cen X, Zhao H. Mitochondrial protein dysfunction in pathogenesis of neurological diseases. Front Mol Neurosci 2022; 15:974480. [PMID: 36157077 PMCID: PMC9489860 DOI: 10.3389/fnmol.2022.974480] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Mitochondria are essential organelles for neuronal function and cell survival. Besides the well-known bioenergetics, additional mitochondrial roles in calcium signaling, lipid biogenesis, regulation of reactive oxygen species, and apoptosis are pivotal in diverse cellular processes. The mitochondrial proteome encompasses about 1,500 proteins encoded by both the nuclear DNA and the maternally inherited mitochondrial DNA. Mutations in the nuclear or mitochondrial genome, or combinations of both, can result in mitochondrial protein deficiencies and mitochondrial malfunction. Therefore, mitochondrial quality control by proteins involved in various surveillance mechanisms is critical for neuronal integrity and viability. Abnormal proteins involved in mitochondrial bioenergetics, dynamics, mitophagy, import machinery, ion channels, and mitochondrial DNA maintenance have been linked to the pathogenesis of a number of neurological diseases. The goal of this review is to give an overview of these pathways and to summarize the interconnections between mitochondrial protein dysfunction and neurological diseases.
Collapse
Affiliation(s)
- Liang Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Ziyun Yang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Xiumei He
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shiming Pu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Cheng Yang
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Qiong Wu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Zuping Zhou
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Hongxia Zhao
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Gangfuß A, Hentschel A, Rademacher N, Sickmann A, Stüve B, Horvath R, Gross C, Kohlschmidt N, Förster F, Abicht A, Schänzer A, Schara-Schmidt U, Roos A, Della Marina A. Identification of a novel homozygous SCO2 variant in siblings with early-onset axonal Charcot-Marie-Tooth disease. Hum Mutat 2022; 43:477-486. [PMID: 35112411 DOI: 10.1002/humu.24338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/23/2022] [Accepted: 01/30/2022] [Indexed: 11/11/2022]
Abstract
The synthesis of cytochrome c oxidase 2 (SCO2) gene encodes for a mitochondrial located metallochaperone essential for the synthesis of the cytochrome c oxidase (COX) subunit 2. Recessive mutations in SCO2 have been reported in several cases with fatal infantile cardioencephalomyopathy with COX deficiency and in only four cases with axonal neuropathy. Here, we identified a homozygous pathogenic variant (c.361G>C; p.(Gly121Arg)) in SCO2 in two brothers with isolated axonal motor neuropathy. To address pathogenicity of the amino acid substitution, biochemical studies were performed and revealed increased level of the mutant SCO2-protein and a dysregulation of COX subunits in leukocytes and moreover unraveled decrease of proteins involved in the manifestation of neuropathies. Hence, our combined data strengthen the concept of SCO2 being causative for a very rare form of axonal neuropathy, expand its molecular genetic spectrum and provide first biochemical insights into the underlying pathophysiology. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Andrea Gangfuß
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - Nina Rademacher
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - Burkhard Stüve
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Rita Horvath
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Claudia Gross
- Institute of Clinical Genetics and Tumor Genetics, Bonn, Germany
| | | | - Fabian Förster
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Angela Abicht
- Department of Neurology, Friedrich-Baur Institute, Munich, Germany.,Medical Genetic Center Munich, Munich, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus Liebig University, Giessen, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Andreas Roos
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany.,Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Adela Della Marina
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
17
|
Rucheton B, Ewenczyk C, Gaignard P, de Sainte Agathe JM, Fauret AL, Saillour V, Leonard-Louis S, Touitou V, Mochel F. Adult Cerebellar Ataxia, Axonal Neuropathy, and Sensory Impairments Caused by Biallelic SCO2 Variants. NEUROLOGY-GENETICS 2021; 7:e630. [PMID: 34746378 PMCID: PMC8569615 DOI: 10.1212/nxg.0000000000000630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/12/2021] [Indexed: 11/15/2022]
Affiliation(s)
- Benoit Rucheton
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Claire Ewenczyk
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Pauline Gaignard
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Jean-Madeleine de Sainte Agathe
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Anne-Laure Fauret
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Virginie Saillour
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Sarah Leonard-Louis
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Valerie Touitou
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Fanny Mochel
- AP-HP (B.R.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle Cardiogénétique et Myogénétique Moléculaire et Cellulaire; AP-HP (C.E., F.M.), Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique; Laboratoire de biologie médicale multisites SeqOIA-FMG2025 (B.R., P.G., J.-M.S.A., A.-L.F., V.S.); AP-HP (P.G.), Université Paris Saclay, CHU Bicêtre, Service de Biochimie, Bicêtre; AP-HP (J.-M.S.A., A.-L.F.), Groupe Hospitalier Pitié-Salpêtrière, Unité Fonctionnelle de Neurogénétique; AP-HP (S.L.-L.), Groupe Hospitalier Pitié-Salpêtrière, Laboratoire de Neuropathologie; AP-HP (V.T.), Groupe Hospitalier Pitié-Salpêtrière Département d'Ophtalmologie, DHU Vision et Handicaps; and Institut du Cerveau (ICM) (F.M.), Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| |
Collapse
|
18
|
Marra F, Lunetti P, Curcio R, Lasorsa FM, Capobianco L, Porcelli V, Dolce V, Fiermonte G, Scarcia P. An Overview of Mitochondrial Protein Defects in Neuromuscular Diseases. Biomolecules 2021; 11:1633. [PMID: 34827632 PMCID: PMC8615828 DOI: 10.3390/biom11111633] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 11/18/2022] Open
Abstract
Neuromuscular diseases (NMDs) are dysfunctions that involve skeletal muscle and cause incorrect communication between the nerves and muscles. The specific causes of NMDs are not well known, but most of them are caused by genetic mutations. NMDs are generally progressive and entail muscle weakness and fatigue. Muscular impairments can differ in onset, severity, prognosis, and phenotype. A multitude of possible injury sites can make diagnosis of NMDs difficult. Mitochondria are crucial for cellular homeostasis and are involved in various metabolic pathways; for this reason, their dysfunction can lead to the development of different pathologies, including NMDs. Most NMDs due to mitochondrial dysfunction have been associated with mutations of genes involved in mitochondrial biogenesis and metabolism. This review is focused on some mitochondrial routes such as the TCA cycle, OXPHOS, and β-oxidation, recently found to be altered in NMDs. Particular attention is given to the alterations found in some genes encoding mitochondrial carriers, proteins of the inner mitochondrial membrane able to exchange metabolites between mitochondria and the cytosol. Briefly, we discuss possible strategies used to diagnose NMDs and therapies able to promote patient outcome.
Collapse
Affiliation(s)
- Federica Marra
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (F.M.); (R.C.); (V.D.)
| | - Paola Lunetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (P.L.); (L.C.)
| | - Rosita Curcio
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (F.M.); (R.C.); (V.D.)
| | - Francesco Massimo Lasorsa
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, via E. Orabona 4, 70125 Bari, Italy; (F.M.L.); (V.P.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, 00155 Rome, Italy
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (P.L.); (L.C.)
| | - Vito Porcelli
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, via E. Orabona 4, 70125 Bari, Italy; (F.M.L.); (V.P.)
| | - Vincenza Dolce
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (F.M.); (R.C.); (V.D.)
| | - Giuseppe Fiermonte
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, via E. Orabona 4, 70125 Bari, Italy; (F.M.L.); (V.P.)
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, 00155 Rome, Italy
| | - Pasquale Scarcia
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, via E. Orabona 4, 70125 Bari, Italy; (F.M.L.); (V.P.)
| |
Collapse
|
19
|
The function of Scox in glial cells is essential for locomotive ability in Drosophila. Sci Rep 2021; 11:21207. [PMID: 34707123 PMCID: PMC8551190 DOI: 10.1038/s41598-021-00663-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/15/2021] [Indexed: 12/19/2022] Open
Abstract
Synthesis of cytochrome c oxidase (Scox) is a Drosophila homolog of human SCO2 encoding a metallochaperone that transports copper to cytochrome c, and is an essential protein for the assembly of cytochrome c oxidase in the mitochondrial respiratory chain complex. SCO2 is highly conserved in a wide variety of species across prokaryotes and eukaryotes, and mutations in SCO2 are known to cause mitochondrial diseases such as fatal infantile cardioencephalomyopathy, Leigh syndrome, and Charcot-Marie-Tooth disease, a neurodegenerative disorder. These diseases have a common symptom of locomotive dysfunction. However, the mechanisms of their pathogenesis remain unknown, and no fundamental medications or therapies have been established for these diseases. In this study, we demonstrated that the glial cell-specific knockdown of Scox perturbs the mitochondrial morphology and function, and locomotive behavior in Drosophila. In addition, the morphology and function of synapses were impaired in the glial cell-specific Scox knockdown. Furthermore, Scox knockdown in ensheathing glia, one type of glial cell in Drosophila, resulted in larval and adult locomotive dysfunction. This study suggests that the impairment of Scox in glial cells in the Drosophila CNS mimics the pathological phenotypes observed by mutations in the SCO2 gene in humans.
Collapse
|
20
|
Gujarati NA, Leonardo AR, Vasquez JM, Guo Y, Frimpong BO, Fozilov E, Revelo MP, Daehn IS, He JC, Bogenhagen D, Mallipattu SK. Loss of Functional SCO2 Attenuates Oxidative Stress in Diabetic Kidney Disease. Diabetes 2021; 71:db210316. [PMID: 34702781 PMCID: PMC8763871 DOI: 10.2337/db21-0316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022]
Abstract
Increased oxidative stress in glomerular endothelial cells (GEnCs) contributes to early diabetic kidney disease (DKD). While mitochondrial respiratory complex IV activity is reduced in DKD, it remains unclear whether this is a driver or a consequence of oxidative stress in GEnCs. Synthesis of cytochrome C oxidase 2 (SCO2), a key metallochaperone in the electron transport chain, is critical to the biogenesis and assembly of subunits required for functional respiratory complex IV activity. Here, we investigated the effects of Sco2 hypomorphs (Sco2 KO/KI , Sco2 KI/KI ), with a functional loss of SCO2, in the progression of DKD using a murine model of Type II Diabetes Mellitus, db/db mice. Diabetic Sco2 KO/KI and Sco2 KI/KI hypomorphs exhibited a reduction in complex IV activity, but an improvement in albuminuria, serum creatinine, and histomorphometric evidence of early DKD as compared to db/db mice. Single-nucleus RNA sequencing with gene set enrichment analysis of differentially expressed genes in the endothelial cluster of Sco2 KO/KI ;db/db mice demonstrated an increase in genes involved in VEGF-VEGFR2 signaling and reduced oxidative stress as compared to db/db mice. These data suggest that reduced complex IV activity due to a loss of functional SCO2 might be protective in GEnCs in early DKD.
Collapse
Affiliation(s)
- Nehaben A Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Alexandra R Leonardo
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Jessica M Vasquez
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Yiqing Guo
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Bismark O Frimpong
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Elbek Fozilov
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Ilse S Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John C He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daniel Bogenhagen
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, NY
- Renal Section, Northport VA Medical Center, Northport, NY
| |
Collapse
|
21
|
Zanfardino P, Doccini S, Santorelli FM, Petruzzella V. Tackling Dysfunction of Mitochondrial Bioenergetics in the Brain. Int J Mol Sci 2021; 22:8325. [PMID: 34361091 PMCID: PMC8348117 DOI: 10.3390/ijms22158325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
Oxidative phosphorylation (OxPhos) is the basic function of mitochondria, although the landscape of mitochondrial functions is continuously growing to include more aspects of cellular homeostasis. Thanks to the application of -omics technologies to the study of the OxPhos system, novel features emerge from the cataloging of novel proteins as mitochondrial thus adding details to the mitochondrial proteome and defining novel metabolic cellular interrelations, especially in the human brain. We focussed on the diversity of bioenergetics demand and different aspects of mitochondrial structure, functions, and dysfunction in the brain. Definition such as 'mitoexome', 'mitoproteome' and 'mitointeractome' have entered the field of 'mitochondrial medicine'. In this context, we reviewed several genetic defects that hamper the last step of aerobic metabolism, mostly involving the nervous tissue as one of the most prominent energy-dependent tissues and, as consequence, as a primary target of mitochondrial dysfunction. The dual genetic origin of the OxPhos complexes is one of the reasons for the complexity of the genotype-phenotype correlation when facing human diseases associated with mitochondrial defects. Such complexity clinically manifests with extremely heterogeneous symptoms, ranging from organ-specific to multisystemic dysfunction with different clinical courses. Finally, we briefly discuss the future directions of the multi-omics study of human brain disorders.
Collapse
Affiliation(s)
- Paola Zanfardino
- Department of Medical Basic Sciences, Neurosciences and Sense Organs, University of Bari Aldo Moro, 70124 Bari, Italy;
| | - Stefano Doccini
- IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy;
| | | | - Vittoria Petruzzella
- Department of Medical Basic Sciences, Neurosciences and Sense Organs, University of Bari Aldo Moro, 70124 Bari, Italy;
| |
Collapse
|
22
|
Huff TC, Sant DW, Camarena V, Van Booven D, Andrade NS, Mustafi S, Monje PV, Wang G. Vitamin C regulates Schwann cell myelination by promoting DNA demethylation of pro-myelinating genes. J Neurochem 2021; 157:1759-1773. [PMID: 32219848 PMCID: PMC7530063 DOI: 10.1111/jnc.15015] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022]
Abstract
Ascorbic acid (vitamin C) is critical for Schwann cells to myelinate peripheral nerve axons during development and remyelination after injury. However, its exact mechanism remains elusive. Vitamin C is a dietary nutrient that was recently discovered to promote active DNA demethylation. Schwann cell myelination is characterized by global DNA demethylation in vivo and may therefore be regulated by vitamin C. We found that vitamin C induces a massive transcriptomic shift (n = 3,848 genes) in primary cultured Schwann cells while simultaneously producing a global increase in genomic 5-hydroxymethylcytosine (5hmC), a DNA demethylation intermediate which regulates transcription. Vitamin C up-regulates 10 pro-myelinating genes which exhibit elevated 5hmC content in both the promoter and gene body regions of these loci following treatment. Using a mouse model of human vitamin C metabolism, we found that maternal dietary vitamin C deficiency causes peripheral nerve hypomyelination throughout early development in resulting offspring. Additionally, dietary vitamin C intake regulates the expression of myelin-related proteins such as periaxin (PRX) and myelin basic protein (MBP) during development and remyelination after injury in mice. Taken together, these results suggest that vitamin C cooperatively promotes myelination through 1) increased DNA demethylation and transcription of pro-myelinating genes, and 2) its known role in stabilizing collagen helices to form the basal lamina that is necessary for myelination.
Collapse
Affiliation(s)
- Tyler C. Huff
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David W. Sant
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vladimir Camarena
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nadja S. Andrade
- Department of Psychiatry & Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sushmita Mustafi
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Paula V. Monje
- Department of Neurological Surgery, Indiana University, Indianapolis, IN, USA
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Hereditary motor neuropathies (HMN) comprise a broad genotypic and phenotypic spectrum of rare, progressively disabling diseases manifesting with length-dependent muscle weakness and atrophy. To date, more than half of the cases cannot be genetically explained. To provide symptomatic and disease-modifying treatments in the future, a better understanding of disease mechanisms is required. RECENT FINDINGS By whole exome and genome sequencing, the discovery of several novel genes (SCO2, TDRKH, SPTAN1, CADM3, and SORD) involved in the pathogenesis of HMN has now relevantly changed the pathophysiological knowledge. This recent success in causative understanding has mainly been driven by the development of functional models including cell culture, animal, and patient-derived induced pluripotent stem cell platforms. These models have an important impact on therapeutic advances including broader approaches to prevent or reverse axonal degeneration and individualized gene silencing attempts using sequence-specific RNA degradation mechanisms. SUMMARY In rare diseases such as HMN, the recent development of genetic sequencing and data interpretation methods has enabled a broader diagnostic approach, whereas treatment strategies are becoming more individualized. Significant milestones have been reached in the discovery of new genes, the establishment of functional disease models, and the preclinical development of mechanistic-based therapies.
Collapse
|
24
|
Dong HL, Ma Y, Yu H, Wei Q, Li JQ, Liu GL, Li HF, Chen L, Chen DF, Bai G, Wu ZY. Bi-allelic loss of function variants in COX20 gene cause autosomal recessive sensory neuronopathy. Brain 2021; 144:2457-2470. [PMID: 33751098 DOI: 10.1093/brain/awab135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/18/2021] [Accepted: 01/30/2021] [Indexed: 12/24/2022] Open
Abstract
Sensory neuronopathies are a rare and distinct subgroup of peripheral neuropathies, characterized by degeneration of the dorsal root ganglia neurons. About 50% of sensory neuronopathies are idiopathic and genetic causes remain to be clarified. Through a combination of homozygosity mapping and whole exome sequencing, we linked an autosomal recessive sensory neuronopathy to pathogenic variants in COX20 gene. We identified 8 unrelated families from the eastern China population carrying a founder variant c.41A>G (p. Lys14Arg) within COX20 in either a homozygous or compound heterozygous state. All patients displayed sensory ataxia with non-length-dependent sensory potentials decrease. COX20 encodes a key transmembrane protein implicated in the assembly of mitochondrial complex IV. We showed that COX20 variants lead to reduction of COX20 protein in patient's fibroblasts and transfected cell lines, consistent with a loss-of-function mechanism. Knockdown of COX20 expression in ND7/23 sensory neuron cells resulted in complex IV deficiency and perturbed assembly of complex IV, which subsequently compromised cell spare respiratory capacity and reduced cell proliferation under metabolic stress. Consistent with mitochondrial dysfunction in knockdown cells, reduced complex IV assembly, enzyme activity and oxygen consumption rate were also found in patients' fibroblasts. We speculated that the mechanism of COX20 was similar to other causative genes (e.g. SURF1, COX6A1, COA3 and SCO2) for peripheral neuropathies, all of which were functionally important in the structure and assembly of complex IV. Our study identifies a novel causative gene for the autosomal recessive sensory neuronopathy, whose vital function in complex IV and high expression in the proprioceptive sensory neuron further underlines loss of COX20 contributing to mitochondrial bioenergetic dysfunction as a mechanism in peripheral sensory neuron disease.
Collapse
Affiliation(s)
- Hai-Lin Dong
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China
| | - Yin Ma
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China
| | - Hao Yu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China
| | - Qiao Wei
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China
| | - Jia-Qi Li
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China
| | - Gong-Lu Liu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China
| | - Hong-Fu Li
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China
| | - Lei Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China.,Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dian-Fu Chen
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China.,Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Ge Bai
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China.,Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Hangzhou, China.,Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China
| |
Collapse
|
25
|
Mitochondrial Structure and Bioenergetics in Normal and Disease Conditions. Int J Mol Sci 2021; 22:ijms22020586. [PMID: 33435522 PMCID: PMC7827222 DOI: 10.3390/ijms22020586] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are ubiquitous intracellular organelles found in almost all eukaryotes and involved in various aspects of cellular life, with a primary role in energy production. The interest in this organelle has grown stronger with the discovery of their link to various pathologies, including cancer, aging and neurodegenerative diseases. Indeed, dysfunctional mitochondria cannot provide the required energy to tissues with a high-energy demand, such as heart, brain and muscles, leading to a large spectrum of clinical phenotypes. Mitochondrial defects are at the origin of a group of clinically heterogeneous pathologies, called mitochondrial diseases, with an incidence of 1 in 5000 live births. Primary mitochondrial diseases are associated with genetic mutations both in nuclear and mitochondrial DNA (mtDNA), affecting genes involved in every aspect of the organelle function. As a consequence, it is difficult to find a common cause for mitochondrial diseases and, subsequently, to offer a precise clinical definition of the pathology. Moreover, the complexity of this condition makes it challenging to identify possible therapies or drug targets.
Collapse
|
26
|
Fernandez-Vizarra E, Zeviani M. Mitochondrial disorders of the OXPHOS system. FEBS Lett 2020; 595:1062-1106. [PMID: 33159691 DOI: 10.1002/1873-3468.13995] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/21/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022]
Abstract
Mitochondrial disorders are among the most frequent inborn errors of metabolism, their primary cause being the dysfunction of the oxidative phosphorylation system (OXPHOS). OXPHOS is composed of the electron transport chain (ETC), formed by four multimeric enzymes and two mobile electron carriers, plus an ATP synthase [also called complex V (cV)]. The ETC performs the redox reactions involved in cellular respiration while generating the proton motive force used by cV to synthesize ATP. OXPHOS biogenesis involves multiple steps, starting from the expression of genes encoded in physically separated genomes, namely the mitochondrial and nuclear DNA, to the coordinated assembly of components and cofactors building each individual complex and eventually the supercomplexes. The genetic cause underlying around half of the diagnosed mitochondrial disease cases is currently known. Many of these cases result from pathogenic variants in genes encoding structural subunits or additional factors directly involved in the assembly of the ETC complexes. Here, we review the historical and most recent findings concerning the clinical phenotypes and the molecular pathological mechanisms underlying this particular group of disorders.
Collapse
Affiliation(s)
- Erika Fernandez-Vizarra
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Massimo Zeviani
- Venetian Institute of Molecular Medicine, Padova, Italy.,Department of Neurosciences, University of Padova, Italy
| |
Collapse
|
27
|
Human Mitochondrial Pathologies of the Respiratory Chain and ATP Synthase: Contributions from Studies of Saccharomyces cerevisiae. Life (Basel) 2020; 10:life10110304. [PMID: 33238568 PMCID: PMC7700678 DOI: 10.3390/life10110304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
The ease with which the unicellular yeast Saccharomyces cerevisiae can be manipulated genetically and biochemically has established this organism as a good model for the study of human mitochondrial diseases. The combined use of biochemical and molecular genetic tools has been instrumental in elucidating the functions of numerous yeast nuclear gene products with human homologs that affect a large number of metabolic and biological processes, including those housed in mitochondria. These include structural and catalytic subunits of enzymes and protein factors that impinge on the biogenesis of the respiratory chain. This article will review what is currently known about the genetics and clinical phenotypes of mitochondrial diseases of the respiratory chain and ATP synthase, with special emphasis on the contribution of information gained from pet mutants with mutations in nuclear genes that impair mitochondrial respiration. Our intent is to provide the yeast mitochondrial specialist with basic knowledge of human mitochondrial pathologies and the human specialist with information on how genes that directly and indirectly affect respiration were identified and characterized in yeast.
Collapse
|
28
|
Lu JQ, Tarnopolsky MA. Mitochondrial neuropathy and neurogenic features in mitochondrial myopathy. Mitochondrion 2020; 56:52-61. [PMID: 33220502 DOI: 10.1016/j.mito.2020.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/25/2020] [Accepted: 11/02/2020] [Indexed: 01/21/2023]
Abstract
Mitochondrial diseases (MIDs) involve multiple organs including peripheral nerves and skeletal muscle. Mitochondrial neuropathy (MN) and mitochondrial myopathy (MM) are commonly associated and linked at the neuromuscular junction (NMJ). Herein we review MN in connection with neurogenic features of MM, and pathological evidence for the involvement of the peripheral nerve and NMJ in MID patients traditionally assumed to have predominantly MM. MN is not uncommon, but still likely under-reported, and muscle biopsies of MM commonly exhibit neurogenic features. Pathological examination remains the gold standard to assess the nerve and muscle changes in patients with MIDs. Ultrastructural studies by electron microscopy are often necessary to fully characterize the pathology of mitochondrial cytopathy in MN and MM.
Collapse
Affiliation(s)
- Jian-Qiang Lu
- Department of Pathology and Molecular Medicine/Neuropathology, McMaster University, Hamilton, Ontario, Canada.
| | - Mark A Tarnopolsky
- Department of Medicine/Neurology, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
29
|
Functions of Cytochrome c oxidase Assembly Factors. Int J Mol Sci 2020; 21:ijms21197254. [PMID: 33008142 PMCID: PMC7582755 DOI: 10.3390/ijms21197254] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022] Open
Abstract
Cytochrome c oxidase is the terminal complex of eukaryotic oxidative phosphorylation in mitochondria. This process couples the reduction of electron carriers during metabolism to the reduction of molecular oxygen to water and translocation of protons from the internal mitochondrial matrix to the inter-membrane space. The electrochemical gradient formed is used to generate chemical energy in the form of adenosine triphosphate to power vital cellular processes. Cytochrome c oxidase and most oxidative phosphorylation complexes are the product of the nuclear and mitochondrial genomes. This poses a series of topological and temporal steps that must be completed to ensure efficient assembly of the functional enzyme. Many assembly factors have evolved to perform these steps for insertion of protein into the inner mitochondrial membrane, maturation of the polypeptide, incorporation of co-factors and prosthetic groups and to regulate this process. Much of the information about each of these assembly factors has been gleaned from use of the single cell eukaryote Saccharomyces cerevisiae and also mutations responsible for human disease. This review will focus on the assembly factors of cytochrome c oxidase to highlight some of the outstanding questions in the assembly of this vital enzyme complex.
Collapse
|
30
|
Soma S, Morgada MN, Naik MT, Boulet A, Roesler AA, Dziuba N, Ghosh A, Yu Q, Lindahl PA, Ames JB, Leary SC, Vila AJ, Gohil VM. COA6 Is Structurally Tuned to Function as a Thiol-Disulfide Oxidoreductase in Copper Delivery to Mitochondrial Cytochrome c Oxidase. Cell Rep 2020; 29:4114-4126.e5. [PMID: 31851937 PMCID: PMC6946597 DOI: 10.1016/j.celrep.2019.11.054] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/07/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022] Open
Abstract
In eukaryotes, cellular respiration is driven by mitochondrial cytochrome c oxidase (CcO), an enzyme complex that requires copper cofactors for its catalytic activity. Insertion of copper into its catalytically active subunits, including COX2, is a complex process that requires metallochaperones and redox proteins including SCO1, SCO2, and COA6, a recently discovered protein whose molecular function is unknown. To uncover the molecular mechanism by which COA6 and SCO proteins mediate copper delivery to COX2, we have solved the solution structure of COA6, which reveals a coiled-coil-helix-coiled-coil-helix domain typical of redox-active proteins found in the mitochondrial inter-membrane space. Accordingly, we demonstrate that COA6 can reduce the copper-coordinating disulfides of its client proteins, SCO1 and COX2, allowing for copper binding. Finally, our determination of the interaction surfaces and reduction potentials of COA6 and its client proteins provides a mechanism of how metallochaperone and disulfide reductase activities are coordinated to deliver copper to CcO. Soma et al. reports the solution structure of cytochrome c oxidase assembly factor COA6 and establishes that it functions as a thiol-disulfide oxidoreductase in a relay system that delivers copper to COX2, a copper-containing subunit of the mitochondrial cytochrome c oxidase.
Collapse
Affiliation(s)
- Shivatheja Soma
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Marcos N Morgada
- Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Área Biofísica, Departamento de Química Biológica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario (2000), Argentina
| | - Mandar T Naik
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Aren Boulet
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Anna A Roesler
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Nathaniel Dziuba
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Alok Ghosh
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA
| | - Qinhong Yu
- Department of Chemistry, University of California, Davis, Davis, CA 95616, USA
| | - Paul A Lindahl
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA; Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - James B Ames
- Department of Chemistry, University of California, Davis, Davis, CA 95616, USA
| | - Scot C Leary
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Alejandro J Vila
- Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET), Área Biofísica, Departamento de Química Biológica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario (2000), Argentina
| | - Vishal M Gohil
- Department of Biochemistry and Biophysics, MS 3474, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
31
|
Inhibition of aggregation and toxicity of α-synuclein in the presence of copper by an N-methylated peptide. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
32
|
Abstract
Mitochondrial disease presenting in childhood is characterized by clinical, biochemical and genetic complexity. Some children are affected by canonical syndromes, but the majority have nonclassical multisystemic disease presentations involving virtually any organ in the body. Each child has a unique constellation of clinical features and disease trajectory, leading to enormous challenges in diagnosis and management of these heterogeneous disorders. This review discusses the classical mitochondrial syndromes presenting most frequently in childhood and then presents an organ-based perspective including systems less frequently linked to mitochondrial disease, such as skin and hair abnormalities and immune dysfunction. An approach to diagnosis is then presented, encompassing clinical evaluation and biochemical, neuroimaging and genetic investigations, and emphasizing the problem of phenocopies. The impact of next-generation sequencing is discussed, together with the importance of functional validation of novel genetic variants never previously linked to mitochondrial disease. The review concludes with a brief discussion of currently available and emerging therapies. The field of mitochondrial medicine has made enormous strides in the last 30 years, with approaching 400 different genes across two genomes now linked to primary mitochondrial disease. However, many important questions remain unanswered, including the reasons for tissue specificity and variability of clinical presentation of individuals sharing identical gene defects, and a lack of disease-modifying therapies and biomarkers to monitor disease progression and/or response to treatment.
Collapse
Affiliation(s)
- S Rahman
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Charcot-Marie-Tooth (CMT) disease and related disorders are the commonest group of inherited neuromuscular diseases and represent a heterogeneous group of disorders. This review will cover recent advances in genetic diagnosis and the evolving genetic and phenotype landscape of this disease group. We will review recent evidence of the increasingly recognized phenotypic overlap with other neurodegenerative conditions including hereditary spastic paraplegia, hereditary ataxias and mitochondrial diseases and highlight the importance of deep phenotyping to inform genetic diagnosis and prognosis. RECENT FINDINGS Through whole exome sequencing and multicentre collaboration new genes are being identified as causal for CMT expanding the genetic heterogeneity of this condition. In addition, an increasing number of variants have been identified in genes known to cause complex inherited diseases in which the peripheral neuropathy is part of the disorder and may be the presenting feature. The recent discovery of a repeat expansion in the RFC1 gene in cerebellar ataxia, neuropathy, vestibular areflexia syndrome highlights the prevalence of late-onset recessive conditions which have historically been considered to cause early-onset disease. SUMMARY CMT is an evolving field with considerable phenotypic and genetic heterogeneity and deep phenotyping remains a cornerstone in contemporary CMT diagnostics.
Collapse
|
34
|
Soh MS, Cheng X, Vijayaraghavan T, Vernon A, Liu J, Neumann B. Disruption of genes associated with Charcot-Marie-Tooth type 2 lead to common behavioural, cellular and molecular defects in Caenorhabditis elegans. PLoS One 2020; 15:e0231600. [PMID: 32294113 PMCID: PMC7159224 DOI: 10.1371/journal.pone.0231600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/26/2020] [Indexed: 11/23/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is an inherited peripheral motor and sensory neuropathy. The disease is divided into demyelinating (CMT1) and axonal (CMT2) neuropathies, and although we have gained molecular information into the details of CMT1 pathology, much less is known about CMT2. Due to its clinical and genetic heterogeneity, coupled with a lack of animal models, common underlying mechanisms remain elusive. In order to gain an understanding of the normal function of genes associated with CMT2, and to draw direct comparisons between them, we have studied the behavioural, cellular and molecular consequences of mutating nine different genes in the nematode Caenorhabditis elegans (lin-41/TRIM2, dyn-1/DNM2, unc-116/KIF5A, fzo-1/MFN2, osm-9/TRPV4, cua-1/ATP7A, hsp-25/HSPB1, hint-1/HINT1, nep-2/MME). We show that C. elegans defective for these genes display debilitated movement in crawling and swimming assays. Severe morphological defects in cholinergic motors neurons are also evident in two of the mutants (dyn-1 and unc-116). Furthermore, we establish methods for quantifying muscle morphology and use these to demonstrate that loss of muscle structure occurs in the majority of mutants studied. Finally, using electrophysiological recordings of neuromuscular junction (NMJ) activity, we uncover reductions in spontaneous postsynaptic current frequency in lin-41, dyn-1, unc-116 and fzo-1 mutants. By comparing the consequences of mutating numerous CMT2-related genes, this study reveals common deficits in muscle structure and function, as well as NMJ signalling when these genes are disrupted.
Collapse
Affiliation(s)
- Ming S. Soh
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Xinran Cheng
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Tarika Vijayaraghavan
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Arwen Vernon
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Jie Liu
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Brent Neumann
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
35
|
Perez-Siles G, Cutrupi A, Ellis M, Kuriakose J, La Fontaine S, Mao D, Uesugi M, Takata RI, Speck-Martins CE, Nicholson G, Kennerson ML. Modelling the pathogenesis of X-linked distal hereditary motor neuropathy using patient-derived iPSCs. Dis Model Mech 2020; 13:13/2/dmm041541. [PMID: 31969342 PMCID: PMC6994953 DOI: 10.1242/dmm.041541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/26/2019] [Indexed: 11/20/2022] Open
Abstract
ATP7A encodes a copper-transporting P-type ATPase and is one of 23 genes in which mutations produce distal hereditary motor neuropathy (dHMN), a group of diseases characterized by length-dependent axonal degeneration of motor neurons. We have generated induced pluripotent stem cell (iPSC)-derived motor neurons from a patient with the p.T994I ATP7A gene mutation as an in vitro model for X-linked dHMN (dHMNX). Patient motor neurons show a marked reduction of ATP7A protein levels in the soma when compared to control motor neurons and failed to upregulate expression of ATP7A under copper-loading conditions. These results recapitulate previous findings obtained in dHMNX patient fibroblasts and in primary cells from a rodent model of dHMNX, indicating that patient iPSC-derived motor neurons will be an important resource for studying the role of copper in the pathogenic processes that lead to axonal degeneration in dHMNX.
Collapse
Affiliation(s)
- Gonzalo Perez-Siles
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, 2139 NSW, Australia .,Sydney Medical School, University of Sydney, Sydney, 2050 NSW, Australia
| | - Anthony Cutrupi
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, 2139 NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, 2050 NSW, Australia
| | - Melina Ellis
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, 2139 NSW, Australia
| | - Jakob Kuriakose
- School of Life Sciences, University of Technology Sydney, Sydney, 2007 NSW, Australia
| | - Sharon La Fontaine
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, 3125 VIC, Australia
| | - Di Mao
- Institute for Integrated Cell-Material Sciences and Institute for Chemical Research, Kyoto University, Kyoto 606-8302, Japan
| | - Motonari Uesugi
- Institute for Integrated Cell-Material Sciences and Institute for Chemical Research, Kyoto University, Kyoto 606-8302, Japan
| | - Reinaldo I Takata
- Sarah Network Rehabilitation Hospitals, Brasilia, 70297-400 DF, Brazil
| | | | - Garth Nicholson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, 2139 NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, 2050 NSW, Australia.,Molecular Medicine Laboratory, Concord Repatriation General Hospital, Sydney, 2139 NSW, Australia
| | - Marina L Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, 2139 NSW, Australia .,Sydney Medical School, University of Sydney, Sydney, 2050 NSW, Australia.,Molecular Medicine Laboratory, Concord Repatriation General Hospital, Sydney, 2139 NSW, Australia
| |
Collapse
|
36
|
Barcia G, Assouline Z, Pennisi A, Gitiaux C, Schiff M, Boddaert N, Munnich A, Bonnefont JP, Rötig A. Cytochrome c oxidase deficiency caused by biallelic SCO2 mutations in two sibs with cerebellar ataxia and progressive peripheral axonal neuropathy. Mol Genet Metab Rep 2019; 21:100528. [PMID: 31844624 PMCID: PMC6895674 DOI: 10.1016/j.ymgmr.2019.100528] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 11/29/2022] Open
Affiliation(s)
- Giulia Barcia
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Zahra Assouline
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Alessandra Pennisi
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Cyril Gitiaux
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Manuel Schiff
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Nathalie Boddaert
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Arnold Munnich
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Jean-Paul Bonnefont
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| | - Agnès Rötig
- Fédération de Génétique Médicale, Hôpital Necker Enfants Malades, Paris, France.,UMR1163, Université Paris Descartes, Sorbonne Paris Cité, Institut IMAGINE, Paris, France
| |
Collapse
|
37
|
Next-generation sequencing in Charcot-Marie-Tooth disease: opportunities and challenges. Nat Rev Neurol 2019; 15:644-656. [PMID: 31582811 DOI: 10.1038/s41582-019-0254-5] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2019] [Indexed: 01/08/2023]
Abstract
Charcot-Marie-Tooth disease and the related disorders hereditary motor neuropathy and hereditary sensory neuropathy, collectively termed CMT, are the commonest group of inherited neuromuscular diseases, and they exhibit wide phenotypic and genetic heterogeneity. CMT is usually characterized by distal muscle atrophy, often with foot deformity, weakness and sensory loss. In the past decade, next-generation sequencing (NGS) technologies have revolutionized genomic medicine and, as these technologies are being applied to clinical practice, they are changing our diagnostic approach to CMT. In this Review, we discuss the application of NGS technologies, including disease-specific gene panels, whole-exome sequencing, whole-genome sequencing (WGS), mitochondrial sequencing and high-throughput transcriptome sequencing, to the diagnosis of CMT. We discuss the growing challenge of variant interpretation and consider how the clinical phenotype can be combined with genetic, bioinformatic and functional evidence to assess the pathogenicity of genetic variants in patients with CMT. WGS has several advantages over the other techniques that we discuss, which include unparalleled coverage of coding, non-coding and intergenic areas of both nuclear and mitochondrial genomes, the ability to identify structural variants and the opportunity to perform genome-wide dense homozygosity mapping. We propose an algorithm for incorporating WGS into the CMT diagnostic pathway.
Collapse
|
38
|
Gualandi F, Sette E, Fortunato F, Bigoni S, De Grandis D, Scotton C, Selvatici R, Neri M, Incensi A, Liguori R, Storbeck M, Karakaya M, Simioni V, Squarzoni S, Timmerman V, Wirth B, Donadio V, Tugnoli V, Ferlini A. Report of a novel ATP7A mutation causing distal motor neuropathy. Neuromuscul Disord 2019; 29:776-785. [DOI: 10.1016/j.nmd.2019.08.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/09/2019] [Accepted: 08/20/2019] [Indexed: 01/26/2023]
|
39
|
Bis-Brewer DM, Fazal S, Züchner S. Genetic modifiers and non-Mendelian aspects of CMT. Brain Res 2019; 1726:146459. [PMID: 31525351 DOI: 10.1016/j.brainres.2019.146459] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/04/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022]
Abstract
Charcot-Marie-Tooth (CMT) neuropathies are amongst the most common inherited diseases in neurology. While great strides have been made to identify the genesis of these diseases, a diagnostic gap of 30-60% remains. Classic models of genetic causation may be limited to fully close this gap and, thus, we review the current state and future role of alternative, non-Mendelian forms of genetics in CMT. Promising synergies exist to further define the full genetic architecture of inherited neuropathies, including affordable whole-genome sequencing, increased data aggregation and clinical collaboration, improved bioinformatics and statistical methodology, and vastly improved computational resources. Given the recent advances in genetic therapies for rare diseases, it becomes a matter of urgency to diagnose CMT patients with great fidelity. Otherwise, they will not be able to benefit from such therapeutic options, or worse, suffer harm when pathogenicity of genetic variation is falsely evaluated. In addition, the newly identified modifier and risk genes may offer alternative targets for pharmacotherapy of inherited and, potentially, even acquired forms of neuropathies.
Collapse
Affiliation(s)
- Dana M Bis-Brewer
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sarah Fazal
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation, Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
40
|
Shy ME. Reply: The classification of Charcot-Marie-Tooth diseases, a never-ending story: CMT4? Brain 2019; 141:e71. [PMID: 30084871 DOI: 10.1093/brain/awy208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Michael E Shy
- Department of Neurology, Carver College of Medicine, University of Iowa, 200 Hawkins Drive, Iowa City Iowa, USA
| |
Collapse
|
41
|
Observation of novel COX20 mutations related to autosomal recessive axonal neuropathy and static encephalopathy. Hum Genet 2019; 138:749-756. [DOI: 10.1007/s00439-019-02026-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023]
|
42
|
Relapsing-Remitting Course of Cystic Leukoencephalopathy. Pediatr Neurol 2018; 89:63-65. [PMID: 30340907 DOI: 10.1016/j.pediatrneurol.2018.08.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/06/2018] [Accepted: 08/18/2018] [Indexed: 11/21/2022]
|
43
|
McCormick EM, Zolkipli-Cunningham Z, Falk MJ. Mitochondrial disease genetics update: recent insights into the molecular diagnosis and expanding phenotype of primary mitochondrial disease. Curr Opin Pediatr 2018; 30:714-724. [PMID: 30199403 PMCID: PMC6467265 DOI: 10.1097/mop.0000000000000686] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Primary mitochondrial disease (PMD) is a genetically and phenotypically diverse group of inherited energy deficiency disorders caused by impaired mitochondrial oxidative phosphorylation (OXPHOS) capacity. Mutations in more than 350 genes in both mitochondrial and nuclear genomes are now recognized to cause primary mitochondrial disease following every inheritance pattern. Next-generation sequencing technologies have dramatically accelerated mitochondrial disease gene discovery and diagnostic yield. Here, we provide an up-to-date review of recently identified, novel mitochondrial disease genes and/or pathogenic variants that directly impair mitochondrial structure, dynamics, and/or function. RECENT FINDINGS A review of PubMed publications was performed from the past 12 months that identified 16 new PMD genes and/or pathogenic variants, and recognition of expanded phenotypes for a wide variety of mitochondrial disease genes. SUMMARY Broad-based exome sequencing has become the standard first-line diagnostic approach for PMD. This has facilitated more rapid and accurate disease identification, and greatly expanded understanding of the wide spectrum of potential clinical phenotypes. A comprehensive dual-genome sequencing approach to PMD diagnosis continues to improve diagnostic yield, advance understanding of mitochondrial physiology, and provide strong potential to develop precision therapeutics targeted to diverse aspects of mitochondrial disease pathophysiology.
Collapse
Affiliation(s)
- Elizabeth M. McCormick
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, PA 19104
| | - Zarazuela Zolkipli-Cunningham
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, PA 19104
| | - Marni J. Falk
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, PA 19104
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
44
|
Human diseases associated with defects in assembly of OXPHOS complexes. Essays Biochem 2018; 62:271-286. [PMID: 30030362 PMCID: PMC6056716 DOI: 10.1042/ebc20170099] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/13/2018] [Accepted: 05/02/2018] [Indexed: 02/02/2023]
Abstract
The structural biogenesis and functional proficiency of the multiheteromeric complexes forming the mitochondrial oxidative phosphorylation system (OXPHOS) require the concerted action of a number of chaperones and other assembly factors, most of which are specific for each complex. Mutations in a large number of these assembly factors are responsible for mitochondrial disorders, in most cases of infantile onset, typically characterized by biochemical defects of single specific complexes. In fact, pathogenic mutations in complex-specific assembly factors outnumber, in many cases, the repertoire of mutations found in structural subunits of specific complexes. The identification of patients with specific defects in assembly factors has provided an important contribution to the nosological characterization of mitochondrial disorders, and has also been a crucial means to identify a huge number of these proteins in humans, which play an essential role in mitochondrial bioenergetics. The wide use of next generation sequencing (NGS) has led to and will allow the identifcation of additional components of the assembly machinery of individual complexes, mutations of which are responsible for human disorders. The functional studies on patients' specimens, together with the creation and characterization of in vivo models, are fundamental to better understand the mechanisms of each of them. A new chapter in this field will be, in the near future, the discovery of mechanisms and actions underlying the formation of supercomplexes, molecular structures formed by the physical, and possibly functional, interaction of some of the individual respiratory complexes, particularly complex I (CI), III (CIII), and IV (CIV).
Collapse
|