1
|
Zhu F, Gao C, Zhu X, Jiang H, Huang M, Zhou Y. Case Report: Charcot-marie-tooth disease caused by a de novo MORC2 gene mutation - novel insights into pathogenicity and treatment. Front Genet 2024; 15:1400906. [PMID: 39464795 PMCID: PMC11512448 DOI: 10.3389/fgene.2024.1400906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/27/2024] [Indexed: 10/29/2024] Open
Abstract
Charcot-Marie-Tooth disease (CMT) is a hereditary peripheral neuropathy involving approximately 80 pathogenic genes. Whole-exome sequencing (WES) and confirmatory Sanger sequencing analysis was applied to identify the disease-causing mutations in a Chinese patient with lower limb weakness. We present an 18-year-old male with a 2.5-year history of progressive lower limb weakness and an unsteady gait. Upon admission, a physical examination revealed hands tremulousness, bilateral calf muscle wasting and weakness, pes cavus, and elevated serum creatine kinase (CK) levels. Electromyography demonstrated axonal neuropathy affecting both upper and lower limbs. A de novo heterozygous missense mutation was identified in the MORC2 gene, NM_001303256.3: c.1199A>G, NP_001290186.1: p.Gln400Arg. Consequently, these clinical and genetic findings suggested a diagnosis of hereditary peripheral neuropathy, CMT type 2Z. Oral mecobalamin and coenzyme Q10 was initiated as subsequent treatment. Our study firstly reports the MORC2 c.1199A>G mutation occurring de novo, highlighting its causal association with CMT2Z, and prompting its reclassification as likely pathogenic. Oral mecobalamin and coenzyme Q10 might be a potential treatment approach for early-stage CMT2Z. We recommend genetic testing for CMT patients to identify the genetic etiology, thereby improving clinical management and facilitating genetic counseling.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Chengcheng Gao
- Zhejiang Key Laboratory of Digital Technology in Medical Diagnostics, Dian Diagnostics Group Co., Ltd., Hangzhou, China
| | - Xiangxiang Zhu
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Huihua Jiang
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Mingchun Huang
- Supply-Room, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Yuanlin Zhou
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| |
Collapse
|
2
|
Hanada K, Osaki Y, Miyamoto R, Muto K, Haji S, Nazere K, Kuwano Y, Morino H, Azuma Y, Miyatake S, Matsumoto N, Izumi Y. Intermediate phenotype between CMT2Z and DIGFAN associated with a novel MORC2 variant: a case report. Hum Genome Var 2024; 11:29. [PMID: 39143067 PMCID: PMC11324651 DOI: 10.1038/s41439-024-00287-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Charcot-Marie-Tooth disease type 2Z is caused by MORC2 mutations and presents with axonal neuropathy. MORC2 mutations can also manifest as developmental delay, impaired growth, dysmorphic facies, and axonal neuropathy (DIGFAN). We report a patient exhibiting an intermediate phenotype between these diseases associated with a novel MORC2 variant. A literature review revealed that the genotype‒phenotype correlation in MORC2-related disorders is complex and that the same mutation can cause a variety of phenotypes.
Collapse
Affiliation(s)
- Kenta Hanada
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.
- Naka Municipal Kaminaka Hospital, Naka, Japan.
| | - Yusuke Osaki
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ryosuke Miyamoto
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kohei Muto
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shotaro Haji
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keyoumu Nazere
- Department of Medical Genetics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuki Kuwano
- Department of Medical Genetics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiroyuki Morino
- Department of Medical Genetics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoshiteru Azuma
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
3
|
Mohapatra B, Pakala SB. Emerging roles of the chromatin remodeler MORC2 in cancer metabolism. Med Oncol 2024; 41:221. [PMID: 39117768 DOI: 10.1007/s12032-024-02464-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
Cancer is characterized by metabolic reprogramming in cancer cells, which is crucial for tumorigenesis. The highly deregulated chromatin remodeler MORC2 contributes to cell proliferation, invasion, migration, DNA repair, and chemoresistance. MORC2 also plays a key role in metabolic reprogramming, including lipogenesis, glucose, and glutamine metabolism. A recent study showed that MORC2-regulated glucose metabolism affects the expression of E-cadherin, a crucial protein in the epithelial-to-mesenchymal transition. This review discusses recent developments in MORC2 regulated cancer cell metabolism and its role in cancer progression.
Collapse
Affiliation(s)
- Bibhukalyan Mohapatra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, India
| | - Suresh B Pakala
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500 046, India.
| |
Collapse
|
4
|
Chung HY, Lee GS, Nam SH, Lee JH, Han JP, Song S, Kim GD, Jung C, Hyeon DY, Hwang D, Choi BO, Yeom SC. Morc2a variants cause hydroxyl radical-mediated neuropathy and are rescued by restoring GHKL ATPase. Brain 2024; 147:2114-2127. [PMID: 38227798 DOI: 10.1093/brain/awae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
Mutations in the Microrchidia CW-type zinc finger 2 (MORC2) GHKL ATPase module cause a broad range of neuropathies, such as Charcot-Marie-Tooth disease type 2Z; however, the aetiology and therapeutic strategy are not fully understood. Previously, we reported that the Morc2a p.S87L mouse model exhibited neuropathy and muscular dysfunction through DNA damage accumulation. In the present study, we analysed the gene expression of Morc2a p.S87L mice and designated the primary causing factor. We investigated the pathological pathway using Morc2a p.S87L mouse embryonic fibroblasts and human fibroblasts harbouring MORC2 p.R252W. We subsequently assessed the therapeutic effect of gene therapy administered to Morc2a p.S87L mice. This study revealed that Morc2a p.S87L causes a protein synthesis defect, resulting in the loss of function of Morc2a and high cellular apoptosis induced by high hydroxyl radical levels. We considered the Morc2a GHKL ATPase domain as a therapeutic target because it simultaneously complements hydroxyl radical scavenging and ATPase activity. We used the adeno-associated virus (AAV)-PHP.eB serotype, which has a high CNS transduction efficiency, to express Morc2a or Morc2a GHKL ATPase domain protein in vivo. Notably, AAV gene therapy ameliorated neuropathy and muscular dysfunction with a single treatment. Loss-of-function characteristics due to protein synthesis defects in Morc2a p.S87L were also noted in human MORC2 p.S87L or p.R252W variants, indicating the correlation between mouse and human pathogenesis. In summary, CMT2Z is known as an incurable genetic disorder, but the present study demonstrated its mechanisms and treatments based on established animal models. This study demonstrates that the Morc2a p.S87L variant causes hydroxyl radical-mediated neuropathy, which can be rescued through AAV-based gene therapy.
Collapse
Affiliation(s)
- Hye Yoon Chung
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Geon Seong Lee
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Soo Hyun Nam
- Samsung Medical Center, Cell & Gene Therapy Institute, Seoul 06351, Korea
| | - Jeong Hyeon Lee
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Jeong Pil Han
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Sumin Song
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Gap-Don Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Choonkyun Jung
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Do Young Hyeon
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Bioinformatics Institute, Bio-MAX, Seoul National University, Seoul 08826, Republic of Korea
| | - Byung-Ok Choi
- Samsung Medical Center, Cell & Gene Therapy Institute, Seoul 06351, Korea
- Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul 06351, Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
- Department of Agricultural Biotechnology, WCU Biomodulation Major, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
5
|
Martinez-Mayer J, Brinkmeier ML, O'Connell SP, Ukagwu A, Marti MA, Miras M, Forclaz MV, Benzrihen MG, Cheung LYM, Camper SA, Ellsworth BS, Raetzman LT, Pérez-Millán MI, Davis SW. Knockout mice with pituitary malformations help identify human cases of hypopituitarism. Genome Med 2024; 16:75. [PMID: 38822427 PMCID: PMC11140907 DOI: 10.1186/s13073-024-01347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Congenital hypopituitarism (CH) and its associated syndromes, septo-optic dysplasia (SOD) and holoprosencephaly (HPE), are midline defects that cause significant morbidity for affected people. Variants in 67 genes are associated with CH, but a vast majority of CH cases lack a genetic diagnosis. Whole exome and whole genome sequencing of CH patients identifies sequence variants in genes known to cause CH, and in new candidate genes, but many of these are variants of uncertain significance (VUS). METHODS The International Mouse Phenotyping Consortium (IMPC) is an effort to establish gene function by knocking-out all genes in the mouse genome and generating corresponding phenotype data. We used mouse embryonic imaging data generated by the Deciphering Mechanisms of Developmental Disorders (DMDD) project to screen 209 embryonic lethal and sub-viable knockout mouse lines for pituitary malformations. RESULTS Of the 209 knockout mouse lines, we identified 51 that have embryonic pituitary malformations. These genes not only represent new candidates for CH, but also reveal new molecular pathways not previously associated with pituitary organogenesis. We used this list of candidate genes to mine whole exome sequencing data of a cohort of patients with CH, and we identified variants in two unrelated cases for two genes, MORC2 and SETD5, with CH and other syndromic features. CONCLUSIONS The screening and analysis of IMPC phenotyping data provide proof-of-principle that recessive lethal mouse mutants generated by the knockout mouse project are an excellent source of candidate genes for congenital hypopituitarism in children.
Collapse
Affiliation(s)
- Julian Martinez-Mayer
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Sean P O'Connell
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA
| | - Arnold Ukagwu
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Marcelo A Marti
- Instituto de Química Biológica de La Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mirta Miras
- Hospital De Niños de La Santísima Trinidad, Córdoba, Argentina
| | - Maria V Forclaz
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Maria G Benzrihen
- Servicio de Endocrinología, Hospital Posadas, Buenos Aires, Argentina
| | - Leonard Y M Cheung
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
- Department of Physiology and Biophyscis, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, 1241 Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Buffy S Ellsworth
- Department of Physiology, Southern Illinois University, 1135 Lincoln Dr, Carbondale, IL, 62901, USA
| | - Lori T Raetzman
- Department of Molecular and Integrative Physiology, University of Illinois, Champaign-Urbana, Urbana, IL, 61801, USA
| | - Maria I Pérez-Millán
- Institute of Biosciences, Biotechnology and Translational Biology (iB3), University of Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA, Buenos Aires, Argentina.
| | - Shannon W Davis
- Department of Biological Sciences, University of South Carolina, 715 Sumter St., Columbia, SC, 29208, USA.
| |
Collapse
|
6
|
Zeng S, Yang H, Wang B, Xie Y, Xu K, Liu L, Cao W, Liu X, Tang B, Liu M, Zhang R. The MORC2 p.S87L mutation reduces proliferation of pluripotent stem cells derived from a patient with the spinal muscular atrophy-like phenotype by inhibiting proliferation-related signaling pathways. Neural Regen Res 2024; 19:205-211. [PMID: 37488868 PMCID: PMC10479865 DOI: 10.4103/1673-5374.375347] [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: 11/07/2022] [Revised: 03/04/2023] [Accepted: 03/29/2023] [Indexed: 07/26/2023] Open
Abstract
Mutations in the microrchidia CW-type zinc finger protein 2 (MORC2) gene are the causative agent of Charcot-Marie-Tooth disease type 2Z (CMT2Z), and the hotspot mutation p.S87L is associated with a more severe spinal muscular atrophy-like clinical phenotype. The aims of this study were to determine the mechanism of the severe phenotype caused by the MORC2 p.S87L mutation and to explore potential treatment strategies. Epithelial cells were isolated from urine samples from a spinal muscular atrophy (SMA)-like patient (MORC2 p.S87L), a CMT2Z patient (MORC2 p.Q400R), and a healthy control and induced to generate pluripotent stem cells, which were then differentiated into motor neuron precursor cells. Next-generation RNA sequencing followed by KEGG pathway enrichment analysis revealed that differentially expressed genes involved in the PI3K/Akt and MAPK/ERK signaling pathways were enriched in the p.S87L SMA-like patient group and were significantly downregulated in induced pluripotent stem cells. Reduced proliferation was observed in the induced pluripotent stem cells and motor neuron precursor cells derived from the p.S87L SMA-like patient group compared with the CMT2Z patient group and the healthy control. G0/G1 phase cell cycle arrest was observed in induced pluripotent stem cells derived from the p.S87L SMA-like patient. MORC2 p.S87L-specific antisense oligonucleotides (p.S87L-ASO-targeting) showed significant efficacy in improving cell proliferation and activating the PI3K/Akt and MAPK/ERK pathways in induced pluripotent stem cells. However, p.S87L-ASO-targeting did not rescue proliferation of motor neuron precursor cells. These findings suggest that downregulation of the PI3K/Akt and MAPK/ERK signaling pathways leading to reduced cell proliferation and G0/G1 phase cell cycle arrest in induced pluripotent stem cells might be the underlying mechanism of the severe p.S87L SMA-like phenotype. p.S87L-ASO-targeting treatment can alleviate disordered cell proliferation in the early stage of pluripotent stem cell induction.
Collapse
Affiliation(s)
- Sen Zeng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Honglan Yang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Binghao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yongzhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Ke Xu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lei Liu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wanqian Cao
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xionghao Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan Province, China
| | - Beisha Tang
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan Province, China
| | - Mujun Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, China
| | - Ruxu Zhang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
7
|
Liu S, Liu X, Lin X, Chen H. Zinc Finger Proteins in the War on Gastric Cancer: Molecular Mechanism and Clinical Potential. Cells 2023; 12:cells12091314. [PMID: 37174714 PMCID: PMC10177130 DOI: 10.3390/cells12091314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
According to the 2020 global cancer data released by the World Cancer Research Fund (WCRF) International, gastric cancer (GC) is the fifth most common cancer worldwide, with yearly increasing incidence and the second-highest fatality rate in malignancies. Despite the contemporary ambiguous molecular mechanisms in GC pathogenesis, numerous in-depth studies have demonstrated that zinc finger proteins (ZFPs) are essential for the development and progression of GC. ZFPs are a class of transcription factors with finger-like domains that bind to Zn2+ extensively and participate in gene replication, cell differentiation and tumor development. In this review, we briefly outline the roles, molecular mechanisms and the latest advances in ZFPs in GC, including eight principal aspects, such as cell proliferation, epithelial-mesenchymal transition (EMT), invasion and metastasis, inflammation and immune infiltration, apoptosis, cell cycle, DNA methylation, cancer stem cells (CSCs) and drug resistance. Intriguingly, the myeloid zinc finger 1 (MZF1) possesses reversely dual roles in GC by promoting tumor proliferation or impeding cancer progression via apoptosis. Therefore, a thorough understanding of the molecular mechanism of ZFPs on GC progression will pave the solid way for screening the potentially effective diagnostic indicators, prognostic biomarkers and therapeutic targets of GC.
Collapse
Affiliation(s)
- Shujie Liu
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Xingzhu Liu
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Xin Lin
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Hongping Chen
- Department of Histology and Embryology, Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
8
|
Stafki SA, Turner J, Littel HR, Bruels CC, Truong D, Knirsch U, Stettner GM, Graf U, Berger W, Kinali M, Jungbluth H, Pacak CA, Hughes J, Mirchi A, Derksen A, Vincent-Delorme C, Theil AF, Bernard G, Ellis D, Fassihi H, Lehmann AR, Laugel V, Mohammed S, Kang PB. The Spectrum of MORC2-Related Disorders: A Potential Link to Cockayne Syndrome. Pediatr Neurol 2023; 141:79-86. [PMID: 36791574 PMCID: PMC10098370 DOI: 10.1016/j.pediatrneurol.2023.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Cockayne syndrome (CS) is a DNA repair disorder primarily associated with pathogenic variants in ERCC6 and ERCC8. As in other Mendelian disorders, there are a number of genetically unsolved CS cases. METHODS We ascertained five individuals with monoallelic pathogenic variants in MORC2, previously associated with three dominantly inherited phenotypes: an axonal form of Charcot-Marie-Tooth disease type 2Z; a syndrome of developmental delay, impaired growth, dysmorphic facies, and axonal neuropathy; and a rare form of spinal muscular atrophy. RESULTS One of these individuals bore a strong phenotypic resemblance to CS. We then identified monoallelic pathogenic MORC2 variants in three of five genetically unsolved individuals with a clinical diagnosis of CS. In total, we identified eight individuals with MORC2-related disorder, four of whom had clinical features strongly suggestive of CS. CONCLUSIONS Our findings indicate that some forms of MORC2-related disorder have phenotypic similarities to CS, including features of accelerated aging. Unlike classic DNA repair disorders, MORC2-related disorder does not appear to be associated with a defect in transcription-coupled nucleotide excision repair and follows a dominant pattern of inheritance with variants typically arising de novo. Such de novo pathogenic variants present particular challenges with regard to both initial gene discovery and diagnostic evaluations. MORC2 should be included in diagnostic genetic test panels targeting the evaluation of microcephaly and/or suspected DNA repair disorders. Future studies of MORC2 and its protein product, coupled with further phenotypic characterization, will help to optimize the diagnosis, understanding, and therapy of the associated disorders.
Collapse
Affiliation(s)
- Seth A Stafki
- Department of Neurology and Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Johnnie Turner
- Department of Neurology and Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Hannah R Littel
- Department of Neurology and Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Christine C Bruels
- Department of Neurology and Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Don Truong
- Department of Neurology and Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Ursula Knirsch
- Neuromuscular Center Zürich and Department of Pediatric Neurology, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Georg M Stettner
- Neuromuscular Center Zürich and Department of Pediatric Neurology, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Urs Graf
- Institute of Medical Molecular Genetics (IMMG), University of Zürich, Zürich, Switzerland
| | - Wolfgang Berger
- Institute of Medical Molecular Genetics (IMMG), University of Zürich, Zürich, Switzerland; Neuroscience Center Zurich (NCZ), University and ETH Zürich, Zürich, Switzerland; Zürich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland
| | - Maria Kinali
- Department of Brain Sciences, Imperial College London and Portland Hospital HCA International, London, United Kingdom
| | - Heinz Jungbluth
- Evelina Children's Hospital and King's College London, University of Manchester, London, United Kingdom
| | - Christina A Pacak
- Department of Neurology and Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jayne Hughes
- Amy and Friends Cockayne Syndrome/Trichothiodystrophy Support, Wirral, United Kingdom
| | - Amytice Mirchi
- Departments of Neurology and Neurosurgery and Pediatrics, McGill University, Montreal, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Alexa Derksen
- Departments of Neurology and Neurosurgery and Pediatrics, McGill University, Montreal, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | | | - Arjan F Theil
- Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Geneviève Bernard
- Departments of Neurology and Neurosurgery and Pediatrics, McGill University, Montreal, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, Canada; Department of Human Genetics, McGill University, Montreal, Canada; Division of Medical Genetics, Department Specialized Medicine, McGill University Health Center, Montreal, Canada
| | - David Ellis
- South East Genomics Laboratory Hub, Guy's Hospital, London, United Kingdom
| | - Hiva Fassihi
- St. John's Institute of Dermatology, Rare Disease Centre, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alan R Lehmann
- Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom
| | - Vincent Laugel
- Service de Pédiatrie 1, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France; Laboratoire de Génétique médicale, INSERM U1112, Institut de génétique médicale d'Alsace, Faculté de Médecine de Strasbourg, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Shehla Mohammed
- South East Thames Regional Genetics Service and Rare Diseases Centre Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Peter B Kang
- Department of Neurology and Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, Minnesota; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
9
|
Jacquier A, Ribault S, Mendes M, Lacoste N, Risson V, Carras J, Latour P, Nadaj-Pakleza A, Stojkovic T, Schaeffer L. Expanding the phenotypic variability of MORC2 gene mutations: From Charcot-Marie-Tooth disease to late-onset pure motor neuropathy. Hum Mutat 2022; 43:1898-1908. [PMID: 35904125 PMCID: PMC10087860 DOI: 10.1002/humu.24445] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/09/2022] [Accepted: 07/27/2022] [Indexed: 01/25/2023]
Abstract
MORC2 gene encodes a ubiquitously expressed nuclear protein involved in chromatin remodeling, DNA repair, and transcriptional regulation. Heterozygous mutations in MORC2 gene have been associated with a spectrum of disorders affecting the peripheral nervous system such as Charcot-Marie-Tooth (CMT2Z), spinal muscular atrophy-like with or without cerebellar involvement, and a developmental syndrome associated with impaired growth, craniofacial dysmorphism and axonal neuropathy (DIGFAN syndrome). Such variability in clinical manifestations associated with the increasing number of variants of unknown significance detected by next-generation sequencing constitutes a serious diagnostic challenge. Here we report the characterization of an in vitro model to evaluate the pathogenicity of variants of unknown significance based on MORC2 overexpression in a neuroblastoma cell line SH-EP or cortical neurons. Likewise, we show that MORC2 mutants affect survival and trigger apoptosis over time in SH-EP cell line. Furthermore, overexpression in primary cortical neurons increases apoptotic cell death and decreases neurite outgrowth. Altogether, these approaches establish the pathogenicity of two new variants p.Gly444Arg and p.His446Gln in three patients from two families. These new mutations in MORC2 gene are associated with autosomal dominant CMT and with adult late onset proximal motor neuropathy, further increasing the spectrum of clinical manifestations associated with MORC2 mutations.
Collapse
Affiliation(s)
- Arnaud Jacquier
- PGNM, Institut NeuroMyoGène, Université Lyon1-CNRS UMR5261-INSERM U1315, Lyon, France.,Centre de Biotechnologie Cellulaire, CBC Biotec, CHU de Lyon-HCL groupement Est, Bron, France
| | - Shams Ribault
- PGNM, Institut NeuroMyoGène, Université Lyon1-CNRS UMR5261-INSERM U1315, Lyon, France.,Service de Médecine Physique et de Réadaptation, Hôpital Henry Gabrielle, Hospices Civils de Lyon, Saint-Genis-Laval, France
| | - Michel Mendes
- Service de Neurologie, Centro Hospitalar Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Nicolas Lacoste
- PGNM, Institut NeuroMyoGène, Université Lyon1-CNRS UMR5261-INSERM U1315, Lyon, France
| | - Valérie Risson
- PGNM, Institut NeuroMyoGène, Université Lyon1-CNRS UMR5261-INSERM U1315, Lyon, France
| | - Julien Carras
- PGNM, Institut NeuroMyoGène, Université Lyon1-CNRS UMR5261-INSERM U1315, Lyon, France.,Centre de Biotechnologie Cellulaire, CBC Biotec, CHU de Lyon-HCL groupement Est, Bron, France
| | - Philippe Latour
- PGNM, Institut NeuroMyoGène, Université Lyon1-CNRS UMR5261-INSERM U1315, Lyon, France.,Unité fonctionnelle de neurogénétique moléculaire, CHU de Lyon-HCL groupement Est, Bron, France
| | - Aleksandra Nadaj-Pakleza
- Centre de Référence des maladies Neuromusculaires Nord/Est/Ile-de-France, Service de Neurologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Tanya Stojkovic
- Institut de Myologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Laurent Schaeffer
- PGNM, Institut NeuroMyoGène, Université Lyon1-CNRS UMR5261-INSERM U1315, Lyon, France.,Centre de Biotechnologie Cellulaire, CBC Biotec, CHU de Lyon-HCL groupement Est, Bron, France
| |
Collapse
|