1
|
Wang R, Zhang F, Li J, Yang D, Zhao H, Yuan J, Jia Y, Yu W, Guo W, Zou L, Zou K. GATA2 promotes cervical cancer progression under the transcriptional activation of TRIP4. Cell Signal 2025; 132:111778. [PMID: 40180167 DOI: 10.1016/j.cellsig.2025.111778] [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: 10/29/2024] [Revised: 03/09/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The continued rise in recurrence and mortality rates of cervical cancer suggests the need to find novel therapeutic targets. Previous studies suggest that TRIP4 acts as a transcription factor to regulate cervical carcinogenesis and progression. Our aim was to explore whether the key downstream genes of TRIP4 functions same as TRIP4 in promoting cervical cancer development. We analyzed and confirmed the downstream targets of TRIP4 by RNA sequencing in cervical cancer cells with TRIP4 knockdown. The expression correlation between TRIP4 and GATA2 and the effect of GATA2 on cervical cancer cell growth were determined respectively by Western Blot, Scratch, Spheroid, and MTT analyses. Pulldown and ChIP experiments were performed to analyze the binding of TRIP4 to the promoter of GATA2. The clinical significance of GATA2 and TRIP4 expression in cervical cancer patients was analyzed by tissue microarray staining. GATA2 was highly expressed in cervical cancer tissues. Knockdown of GATA2 inhibited the growth, metastasis and stemness of cervical cancer cells and sensitized cervical cancer cells to radiation therapy. The inhibitory effect of TRIP4 knockdown on cervical cancer cells was rescued by GATA2 overexpression. Furthermore, TRIP4 could bind to the specific GATA2 promoter region, thereby activating its transcription. Clinical tissue microarray analysis indicated that the expression of TRIP4 and GATA2 was positively correlated, and high expression of both predicted a poor prognosis in cervical cancer patients. Our study demonstrated that GATA2 functions as the key downstream target of TRIP4 to promote cervical cancer progression and effective intervention of TRIP4/GATA2 signaling is expected to be developed as potential cervical cancer therapeutic strategy.
Collapse
Affiliation(s)
- Ruonan Wang
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Feng Zhang
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiazhi Li
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dian Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Hongmei Zhao
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Yuan
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuhan Jia
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wei Guo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Lijuan Zou
- The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Kun Zou
- The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
2
|
Onnée M, Malfatti E. The widening genetic and myopathologic spectrum of congenital myopathies (CMYOs): a narrative review. Neuromuscul Disord 2025; 49:105338. [PMID: 40112751 DOI: 10.1016/j.nmd.2025.105338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
Congenital myopathies (CMYOs) represent a genetically and clinically heterogeneous group of disorders characterized by early-onset muscle weakness and distinct myopathologic features. The advent of next-generation sequencing (NGS) has accelerated the identification of causative genes, leading to the discovery of novel CMYOs and thereby challenging the traditional classification. In this comprehensive review, we focus on the clinical, myopathologic, molecular and pathophysiological features of 33 newly identified CMYOs.
Collapse
Affiliation(s)
- Marion Onnée
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France
| | - Edoardo Malfatti
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; Assistance Publique-Hôpitaux de Paris, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Filnemus, Hôpital Henri Mondor, 94010 Créteil, France; European Reference Center for Neuromuscular Disorders, EURO-NMD, France.
| |
Collapse
|
3
|
Coppens S, Deconinck N, Sullivan P, Smolnikov A, Clayton JS, Griffin KR, Jones KJ, Vilain CN, Kadhim H, Bryen SJ, Faiz F, Waddell LB, Evesson FJ, Bakshi M, Pinner JR, Charlton A, Brammah S, Graf NS, Krivanek M, Tay CG, Foulds NC, Illingworth MA, Thomas NH, Ellard S, Mazanti I, Park S, French CE, Brewster J, Belteki G, Hoodbhoy S, Allinson K, Krishnakumar D, Baynam G, Wood BM, Ward M, Vijayakumar K, Syed A, Murugan A, Majumdar A, Scurr IJ, Splitt MP, Moldovan C, de Silva DC, Senanayake K, Gardeitchik T, Arens Y, Cooper ST, Laing NG, Raymond FL, Jungbluth H, Kamsteeg E, Manzur A, Corley SM, Ravenscroft G, Wilkins MR, Cowley MJ, Pinese M, Phadke R, Davis MR, Muntoni F, Oates EC. Congenital Titinopathy: Comprehensive Characterization of the Most Severe End of the Disease Spectrum. Ann Neurol 2025; 97:611-628. [PMID: 39853809 PMCID: PMC11889535 DOI: 10.1002/ana.27087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 01/26/2025]
Abstract
Congenital titinopathy has recently emerged as one of the most common congenital muscle disorders. OBJECTIVE To better understand the presentation and clinical needs of the under-characterized extreme end of the congenital titinopathy severity spectrum. METHODS We comprehensively analyzed the clinical, imaging, pathology, autopsy, and genetic findings in 15 severely affected individuals from 11 families. RESULTS Prenatal features included hypokinesia or akinesia and growth restriction. Six pregnancies were terminated. Nine infants were born at or near term with severe-to-profound weakness and required resuscitation. Seven died following withdrawal of life support. Two surviving children require ongoing respiratory support. Most cohort members had at least 1 disease-causing variant predicted to result in some near-normal-length titin expression. The exceptions, from 2 unrelated families, had homozygous truncating variants predicted to induce complete nonsense mediated decay. However, subsequent analyses suggested that the causative variant in each family had an additional previously unrecognized impact on splicing likely to result in some near-normal-length titin expression. This impact was confirmed by minigene assay for 1 variant. INTERPRETATION This study confirms the clinical variability of congenital titinopathy. Severely affected individuals succumb prenatally/during infancy, whereas others survive into adulthood. It is likely that this variability is because of differences in the amount and/or length of expressed titin. If confirmed, analysis of titin expression could facilitate clinical prediction and increasing expression might be an effective treatment strategy. Our findings also further-support the hypothesis that some near-normal-length titin expression is essential to early prenatal survival. Sometimes expression of normal/near-normal-length titin is due to disease-causing variants having an additional impact on splicing. ANN NEUROL 2025;97:611-628.
Collapse
Affiliation(s)
- Sandra Coppens
- Hopital Erasme, ULB Center of Human GeneticsUniversité Libre de BruxellesBrusselsBelgium
| | - Nicolas Deconinck
- Department of Paediatric Neurology, Neuromuscular Reference CenterHôpital Universitaire des Enfants Reine Fabiola, Université Libre de BruxellesBrusselsBelgium
| | - Patricia Sullivan
- Children's Cancer Institute, Lowy Cancer CentreUniversity of New South WalesSydneyNew South WalesAustralia
| | - Andrei Smolnikov
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Joshua S. Clayton
- Harry Perkins Institute of Medical Research, QEII Medical CentreNedlandsWestern AustraliaAustralia
- Centre for Medical ResearchThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Kaitlyn R. Griffin
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Kristi J. Jones
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Catheline N. Vilain
- Hopital Erasme, ULB Center of Human GeneticsUniversité Libre de BruxellesBrusselsBelgium
| | - Hazim Kadhim
- Neuropathology Unit (Anatomic Pathology Service) and Reference Center for Neuromuscular PathologyCHU Brugmann‐HUDERF, Université Libre de BruxellesBrusselsBelgium
| | - Samantha J. Bryen
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Fathimath Faiz
- Department of Diagnostic Genomics, PathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Leigh B. Waddell
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Frances J. Evesson
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
- Functional NeuromicsChildren's Medical Research InstituteWestmeadNew South WalesAustralia
| | - Madhura Bakshi
- Department of Clinical GeneticsLiverpool HospitalLiverpoolNew South WalesAustralia
| | - Jason R. Pinner
- Department of Medical GenomicsRoyal Prince Alfred Hospital, The University of SydneyCamperdownNew South WalesAustralia
| | - Amanda Charlton
- Department of HistopathologyThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Susan Brammah
- Electron Microscope Unit, Department of Anatomical PathologyConcord Repatriation General HospitalConcordNew South WalesAustralia
| | - Nicole S. Graf
- Department of HistopathologyThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Michael Krivanek
- Department of HistopathologyThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
| | - Chee Geap Tay
- Division of Paediatric Neurology, Department of Paediatrics, Faculty of MedicineUniversity of MalayaKuala LumpurMalaysia
| | - Nicola C. Foulds
- Wessex Clinical Genetics ServiceUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Marjorie A. Illingworth
- Department of Paediatric NeurologyUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Neil H. Thomas
- Department of Paediatric NeurologyUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Sian Ellard
- College of Medicine and Health, University of Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation TrustExeterUK
| | - Ingrid Mazanti
- Department of Cellular PathologyUniversity Hospital Southampton NHS Foundation TrustSouthamptonUK
| | - Soo‐Mi Park
- Department of Clinical GeneticsCambridge University Hospital NHS Foundation TrustCambridgeUK
| | - Courtney E. French
- Department of Medical GeneticsCambridge Institute for Medical Research, University of CambridgeCambridgeUK
| | - Jennifer Brewster
- Department of Fetomaternal MedicineCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Gusztav Belteki
- Neonatal Intensive Care UnitCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Shazia Hoodbhoy
- Neonatal Intensive Care UnitCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Kieren Allinson
- Department of HistopathologyCambridge University Hospitals Foundation TrustCambridgeUK
| | - Deepa Krishnakumar
- Department of Paediatric NeurologyCambridge University Hospitals Foundation TrustCambridgeUK
| | - Gareth Baynam
- Genetic Services of Western AustraliaKing Edward Memorial HospitalPerthWestern AustraliaAustralia
| | | | - Michelle Ward
- Genetic Services of Western AustraliaKing Edward Memorial HospitalPerthWestern AustraliaAustralia
| | - Kayal Vijayakumar
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Amber Syed
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Archana Murugan
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Anirban Majumdar
- Department of Paediatric NeurologyUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Ingrid J. Scurr
- Department of Clinical GeneticsUniversity Hospitals Bristol NHS Foundation TrustBristolUK
| | - Miranda P. Splitt
- Northern Genetics ServiceInstitute of Genetic MedicineNewcastle upon TyneUK
| | - Corina Moldovan
- Department of PathologyNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Deepthi C. de Silva
- Department of Physiology, Faculty of MedicineUniversity of KelaniyaRagamaSri Lanka
| | - Kumudu Senanayake
- Department of HistopathologyCastle Street Hospital for WomenColomboSri Lanka
| | - Thatjana Gardeitchik
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | - Yvonne Arens
- Department of Clinical GeneticsMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Sandra T. Cooper
- Kids Neuroscience Centre, Kids ResearchThe Children's Hospital at WestmeadWestmeadNew South WalesAustralia
- Faculty of Medicine and HealthThe University of SydneyWestmeadNew South WalesAustralia
- Functional NeuromicsChildren's Medical Research InstituteWestmeadNew South WalesAustralia
| | - Nigel G. Laing
- Harry Perkins Institute of Medical Research, QEII Medical CentreNedlandsWestern AustraliaAustralia
- Centre for Medical ResearchThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - F. Lucy Raymond
- Department of Medical GeneticsCambridge Institute for Medical Research, University of CambridgeCambridgeUK
| | - Heinz Jungbluth
- Department of Paediatric Neurology, Neuromuscular ServiceEvelina's Children Hospital, Guy's and St. Thomas' Hospital NHS Foundation TrustLondonUK
- Randall Division for Cell and Molecular Biophysics, Muscle Signalling SectionKing's College LondonLondonUK
- Department of Basic and Clinical NeuroscienceIoPPN, King's College LondonLondonUK
| | - Erik‐Jan Kamsteeg
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | - Adnan Manzur
- Great Ormond Street Hospital for ChildrenNHS Foundation Trust, Dubowitz Neuromuscular CentreLondonUK
| | - Susan M. Corley
- Systems Biology Initiative, School of Biotechnology and Biomolecular ScienceUniversity of New South WalesSydneyNew South WalesAustralia
| | - Gianina Ravenscroft
- Harry Perkins Institute of Medical Research, QEII Medical CentreNedlandsWestern AustraliaAustralia
- Centre for Medical ResearchThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Marc R. Wilkins
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Mark J. Cowley
- Children's Cancer Institute, Lowy Cancer CentreUniversity of New South WalesSydneyNew South WalesAustralia
| | - Mark Pinese
- Children's Cancer Institute, Lowy Cancer CentreUniversity of New South WalesSydneyNew South WalesAustralia
| | - Rahul Phadke
- Great Ormond Street Hospital for ChildrenNHS Foundation Trust, Dubowitz Neuromuscular CentreLondonUK
- Division of NeuropathologyUCL Institute of Neurology, The National Hospital for Neurology and NeurosurgeryLondonUK
| | - Mark R. Davis
- Department of Diagnostic Genomics, PathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Francesco Muntoni
- Great Ormond Street Hospital for ChildrenNHS Foundation Trust, Dubowitz Neuromuscular CentreLondonUK
| | - Emily C. Oates
- School of Biotechnology and Biomolecular SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Department of NeurologySydney Children's HospitalSydneyNew South WalesAustralia
| |
Collapse
|
4
|
Wang Y, Zhang M, Zhang T, Zhang S, Ji F, Qin J, Li H, Jiao J. PD-L1/PD-1 checkpoint pathway regulates astrocyte morphogenesis and myelination during brain development. Mol Psychiatry 2025:10.1038/s41380-025-02969-3. [PMID: 40164696 DOI: 10.1038/s41380-025-02969-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/05/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Programmed cell death protein 1 (PD-1) and its primary ligand PD-L1 are integral components of a significant immune checkpoint pathway, widely recognized for its central role in cancer immunotherapy. However, emerging evidence highlights their broader involvement in both the central and peripheral nervous systems. In this study, we demonstrate that PD-L1/PD-1 signaling in astrocytes during mouse brain development regulates astrocyte maturation and morphogenesis via the MEK/ERK pathway by targeting the downstream effector cysteine and glycine rich protein 1 (CSRP1). This enhanced astrocyte morphological complexity results in increased end-foot coverage of blood vessels. Additionally, aberrant secretion of CSRP1 by astrocytes interacts with oligodendrocyte precursor cells (OPCs) membrane proteins annexin A1 (ANXA1) and annexin A2 (ANXA2), leading to the exclusion of migrating OPCs from blood vessels. This disruption in OPC migration and differentiation results in abnormal myelination and is associated with cognitive deficits in the mice. Our results provide critical insights into the function of PD-L1/PD-1 signaling in astrocyte-OPC interactions and underscore its relevance to glial cell development and pathogenesis in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yanyan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengtian Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyu Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shukui Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fen Ji
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Qin
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Li
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianwei Jiao
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
5
|
Nishio H, Niba ETE, Saito T, Okamoto K, Lee T, Takeshima Y, Awano H, Lai PS. Clinical and Genetic Profiles of 5q- and Non-5q-Spinal Muscular Atrophy Diseases in Pediatric Patients. Genes (Basel) 2024; 15:1294. [PMID: 39457418 PMCID: PMC11506990 DOI: 10.3390/genes15101294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is a genetic disease characterized by loss of motor neurons in the spinal cord and lower brainstem. The term "SMA" usually refers to the most common form, 5q-SMA, which is caused by biallelic mutations in SMN1 (located on chromosome 5q13). However, long before the discovery of SMN1, it was known that other forms of SMA existed. Therefore, SMA is currently divided into two groups: 5q-SMA and non-5q-SMA. This is a simple and practical classification, and therapeutic drugs have only been developed for 5q-SMA (nusinersen, onasemnogene abeparvovec, risdiplam) and not for non-5q-SMA disease. METHODS We conducted a non-systematic critical review to identify the characteristics of each SMA disease. RESULTS Many of the non-5q-SMA diseases have similar symptoms, making DNA analysis of patients essential for accurate diagnosis. Currently, genetic analysis technology using next-generation sequencers is rapidly advancing, opening up the possibility of elucidating the pathology and treating non-5q-SMA. CONCLUSION Based on accurate diagnosis and a deeper understanding of the pathology of each disease, treatments for non-5q-SMA diseases may be developed in the near future.
Collapse
Affiliation(s)
- Hisahide Nishio
- Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani-cho, Nishi-ku, Kobe 651-2180, Japan
| | - Emma Tabe Eko Niba
- Laboratory of Molecular and Biochemical Research, Biomedical Research Core Facilities, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Toshio Saito
- Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Toyonaka 560-8552, Japan;
| | - Kentaro Okamoto
- Department of Pediatrics, Ehime Prefectural Imabari Hospital, 4-5-5 Ishi-cho, Imabari 794-0006, Japan;
| | - Tomoko Lee
- Department of Pediatrics, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (T.L.); (Y.T.)
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (T.L.); (Y.T.)
| | - Hiroyuki Awano
- Organization for Research Initiative and Promotion, Research Initiative Center, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan;
| | - Poh-San Lai
- Department of Pediatrics, National University of Singapore, 1E Lower Kent Ridge Road, Singapore 119228, Singapore;
| |
Collapse
|
6
|
Johnson AN. Myotube Guidance: Shaping up the Musculoskeletal System. J Dev Biol 2024; 12:25. [PMID: 39311120 PMCID: PMC11417883 DOI: 10.3390/jdb12030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/20/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Myofibers are highly specialized contractile cells of skeletal muscles, and dysregulation of myofiber morphogenesis is emerging as a contributing cause of myopathies and structural birth defects. Myotubes are the myofiber precursors and undergo a dramatic morphological transition into long bipolar myofibers that are attached to tendons on two ends. Similar to axon growth cones, myotube leading edges navigate toward target cells and form cell-cell connections. The process of myotube guidance connects myotubes with the correct tendons, orients myofiber morphology with the overall body plan, and generates a functional musculoskeletal system. Navigational signaling, addition of mass and volume, and identification of target cells are common events in myotube guidance and axon guidance, but surprisingly, the mechanisms regulating these events are not completely overlapping in myotubes and axons. This review summarizes the strategies that have evolved to direct myotube leading edges to predetermined tendon cells and highlights key differences between myotube guidance and axon guidance. The association of myotube guidance pathways with developmental disorders is also discussed.
Collapse
Affiliation(s)
- Aaron N Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Chinnam NB, Thapar R, Arvai AS, Sarker AH, Soll JM, Paul T, Syed A, Rosenberg DJ, Hammel M, Bacolla A, Katsonis P, Asthana A, Tsai MS, Ivanov I, Lichtarge O, Silverman RH, Mosammaparast N, Tsutakawa SE, Tainer JA. ASCC1 structures and bioinformatics reveal a novel helix-clasp-helix RNA-binding motif linked to a two-histidine phosphodiesterase. J Biol Chem 2024; 300:107368. [PMID: 38750793 PMCID: PMC11214414 DOI: 10.1016/j.jbc.2024.107368] [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: 01/22/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Activating signal co-integrator complex 1 (ASCC1) acts with ASCC-ALKBH3 complex in alkylation damage responses. ASCC1 uniquely combines two evolutionarily ancient domains: nucleotide-binding K-Homology (KH) (associated with regulating splicing, transcriptional, and translation) and two-histidine phosphodiesterase (PDE; associated with hydrolysis of cyclic nucleotide phosphate bonds). Germline mutations link loss of ASCC1 function to spinal muscular atrophy with congenital bone fractures 2 (SMABF2). Herein analysis of The Cancer Genome Atlas (TCGA) suggests ASCC1 RNA overexpression in certain tumors correlates with poor survival, Signatures 29 and 3 mutations, and genetic instability markers. We determined crystal structures of Alvinella pompejana (Ap) ASCC1 and Human (Hs) PDE domain revealing high-resolution details and features conserved over 500 million years of evolution. Extending our understanding of the KH domain Gly-X-X-Gly sequence motif, we define a novel structural Helix-Clasp-Helix (HCH) nucleotide binding motif and show ASCC1 sequence-specific binding to CGCG-containing RNA. The V-shaped PDE nucleotide binding channel has two His-Φ-Ser/Thr-Φ (HXT) motifs (Φ being hydrophobic) positioned to initiate cyclic phosphate bond hydrolysis. A conserved atypical active-site histidine torsion angle implies a novel PDE substrate. Flexible active site loop and arginine-rich domain linker appear regulatory. Small-angle X-ray scattering (SAXS) revealed aligned KH-PDE RNA binding sites with limited flexibility in solution. Quantitative evolutionary bioinformatic analyses of disease and cancer-associated mutations support implied functional roles for RNA binding, phosphodiesterase activity, and regulation. Collective results inform ASCC1's roles in transactivation and alkylation damage responses, its targeting by structure-based inhibitors, and how ASCC1 mutations may impact inherited disease and cancer.
Collapse
Affiliation(s)
- Naga Babu Chinnam
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roopa Thapar
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew S Arvai
- Integrative Structural & Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Altaf H Sarker
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Jennifer M Soll
- Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Tanmoy Paul
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Aleem Syed
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel J Rosenberg
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Michal Hammel
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Albino Bacolla
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Panagiotis Katsonis
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Abhishek Asthana
- Department Cancer Biology, Cleveland Clinic Foundation, Lerner Research Institute, Cleveland, Ohio, USA
| | - Miaw-Sheue Tsai
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Ivaylo Ivanov
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Olivier Lichtarge
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Robert H Silverman
- Department Cancer Biology, Cleveland Clinic Foundation, Lerner Research Institute, Cleveland, Ohio, USA
| | - Nima Mosammaparast
- Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Susan E Tsutakawa
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA.
| | - John A Tainer
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA; Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
8
|
Erdogdu B, Varabyou A, Hicks SC, Salzberg SL, Pertea M. Detecting differential transcript usage in complex diseases with SPIT. CELL REPORTS METHODS 2024; 4:100736. [PMID: 38508189 PMCID: PMC10985272 DOI: 10.1016/j.crmeth.2024.100736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/21/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024]
Abstract
Differential transcript usage (DTU) plays a crucial role in determining how gene expression differs among cells, tissues, and developmental stages, contributing to the complexity and diversity of biological systems. In abnormal cells, it can also lead to deficiencies in protein function and underpin disease pathogenesis. Analyzing DTU via RNA sequencing (RNA-seq) data is vital, but the genetic heterogeneity in populations with complex diseases presents an intricate challenge due to diverse causal events and undetermined subtypes. Although the majority of common diseases in humans are categorized as complex, state-of-the-art DTU analysis methods often overlook this heterogeneity in their models. We therefore developed SPIT, a statistical tool that identifies predominant subgroups in transcript usage within a population along with their distinctive sets of DTU events. This study provides comprehensive assessments of SPIT's methodology and applies it to analyze brain samples from individuals with schizophrenia, revealing previously unreported DTU events in six candidate genes.
Collapse
Affiliation(s)
- Beril Erdogdu
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering, Baltimore, MD, USA.
| | - Ales Varabyou
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering, Baltimore, MD, USA; Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Stephanie C Hicks
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering, Baltimore, MD, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Steven L Salzberg
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering, Baltimore, MD, USA; Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mihaela Pertea
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering, Baltimore, MD, USA; Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Favier M, Delanne J, Gorincour G, Faivre L, Racine C, Philippe C, Duffourd Y, Vitobello A, Rousseau T, Martz O, Tarris G, Oualiken C, Thauvin-Robinet C, Mau-Them FT. Early prenatal diagnosis of causative homozygous variants in ASCC1 in a fetus with cystic hygroma and additional homozygous variants of unknown significance associated with a neurological phenotype not visible in early gestation: Dual diagnosis or not? Prenat Diagn 2024; 44:352-356. [PMID: 38342957 DOI: 10.1002/pd.6519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/23/2023] [Accepted: 12/26/2023] [Indexed: 02/13/2024]
Abstract
A consanguineous couple was referred at 10 weeks of gestation (WG) for prenatal genetic investigations due to isolated cystic hygroma. Prenatal trio exome sequencing identified causative homozygous truncating variants in ASCC1 previously implicated in spinal muscular atrophy with congenital bone fractures. Prenatal manifestations in ASCC1 can usually include hydramnios, fetal hypo-/akinesia, arthrogryposis, contractures and limb deformities, hydrops fetalis and cystic hygroma. An additional truncating variant was identified in CSPP1 associated with Joubert syndrome. Presentations in CSPP1 include cerebellar and brainstem malformations with vermis hypoplasia and molar tooth sign, difficult to visualize in early gestation. A second pregnancy was marked by the recurrence of isolated increased nuchal translucency at 10 + 2 WG. Sanger prenatal diagnosis targeted on ASCC1 and CSPP1 variants showed the presence of the homozygous familial ASCC1 variant. In this case, prenatal exome sequencing analysis is subject to a partial ASCC1 phenotype and an undetectable CSPP1 phenotype at 10 weeks of gestation. As CSPP1 contribution is unclear or speculative to a potentially later in pregnancy or postnatal phenotype, it is mentioned as a variant of uncertain significance. The detection of pathogenic or likely pathogenic variants involved in severe disorders but without phenotype-genotype correlation because the pregnancy is in the early stages or due to prenatally undetectable phenotypes, will encourage the clinical community to define future practices in molecular prenatal reporting.
Collapse
Affiliation(s)
- Maud Favier
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- SoFFoet - Société Française de Foetopathologie, Paris, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Julian Delanne
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Centre Pluridisciplinaire de Diagnostic Prénatal, CHU Dijon Bourgogne, Dijon, France
| | - Guillaume Gorincour
- Institut Méditerranéen d'Imagerie Médicale Appliquée à la Gynécologie, la Grossesse et l'Enfance (IMAGE 2), Marseille, France
| | - Laurence Faivre
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Caroline Racine
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Centre Pluridisciplinaire de Diagnostic Prénatal, CHU Dijon Bourgogne, Dijon, France
| | - Christophe Philippe
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Laboratoire de Génétique, CHR Metz Thionville, Hôpital Mercy, Metz, France
| | - Yannis Duffourd
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Antonio Vitobello
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Thierry Rousseau
- Centre Pluridisciplinaire de Diagnostic Prénatal, CHU Dijon Bourgogne, Dijon, France
| | - Olivia Martz
- Centre Pluridisciplinaire de Diagnostic Prénatal, CHU Dijon Bourgogne, Dijon, France
| | - Georges Tarris
- UMR1098, Service de Pathologie, Université Bourgogne-Franche Comté, Dijon, France
| | - Camélia Oualiken
- UMR1098, Service de Pathologie, Université Bourgogne-Franche Comté, Dijon, France
| | - Christel Thauvin-Robinet
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Centre Pluridisciplinaire de Diagnostic Prénatal, CHU Dijon Bourgogne, Dijon, France
| | - Frédéric Tran Mau-Them
- Inserm UMR1231 - GAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Maladies Rares Anomalies du développement et Syndromes malformatifs, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| |
Collapse
|
10
|
Decio A, Giorda R, Panzeri E, Bassi MT, D'Angelo MG. Clinical phenotype and next-generation sequencing as essential tools for the diagnosis of a rare form of congenital myopathy due to a TRIP4 intragenic deletion. Neurol Sci 2024; 45:819-823. [PMID: 37792112 DOI: 10.1007/s10072-023-07102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023]
Affiliation(s)
- Alice Decio
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute IRCCS E. Medea, Via Don L. Monza 20, 23842, Bosisio Parini (LC), Italy.
| | - Roberto Giorda
- Molecular Biology Laboratory, IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Elena Panzeri
- Molecular Biology Laboratory, IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Maria Teresa Bassi
- Molecular Biology Laboratory, IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Maria Grazia D'Angelo
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute IRCCS E. Medea, Via Don L. Monza 20, 23842, Bosisio Parini (LC), Italy
| |
Collapse
|
11
|
Frongia I, Spagnoli C, Rizzi S, Frattini D, Leon A, Caraffi SG, Pollazzon M, Garavelli L, Pisani F, Fusco C. 'A novel TRIP4 Variant Associated with Peripheral Neuropathy: Expanding the Clinical and Genetic Spectrum of ASC1-Related Myopathy'. J Neuromuscul Dis 2024; 11:213-219. [PMID: 38143368 PMCID: PMC10789366 DOI: 10.3233/jnd-230110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2023] [Indexed: 12/26/2023]
Abstract
Activating Signal Cointegrator 1 complex (ASC-1 complex) is a ribonucleoprotein tetramer participating in transcriptional coactivation and RNA processing, consisting of four subunits: ASCC1-ASCC3 and ASC-1. Pathogenic variants in the TRIP4 and ASCC1 genes, encoding the ASC-1 and ASCC1 subunits, were recently described in congenital myopathic conditions without signs of motor neuron involvement, and Spinal Muscular Atrophy-like (SMA-like) phenotype with prenatal bone fractures. We present a novel pathogenic TRIP4 variant in two siblings with severe phenotype and mixed sensory-motor polyneuropathy. The reviewed phenotypic spectrum is broad, but sensory-motor polyneuropathy is so-far unreported. We thus expand ASC-1 related myopathy phenotype.
Collapse
Affiliation(s)
- Ivana Frongia
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlotta Spagnoli
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Susanna Rizzi
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Daniele Frattini
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Stefano Giuseppe Caraffi
- Struttura Complessa di Genetica Medica, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marzia Pollazzon
- Struttura Complessa di Genetica Medica, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Livia Garavelli
- Struttura Complessa di Genetica Medica, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesco Pisani
- Child Neuropsychiatric Unit, Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Carlo Fusco
- Struttura Complessa di Neuropsichiatria Infantile, Dipartimento Materno-Infantile, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
12
|
Erdogdu B, Varabyou A, Hicks SC, Salzberg SL, Pertea M. Detecting differential transcript usage in complex diseases with SPIT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.10.548289. [PMID: 37503064 PMCID: PMC10369883 DOI: 10.1101/2023.07.10.548289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Differential transcript usage (DTU) plays a crucial role in determining how gene expression differs among cells, tissues, and different developmental stages, thereby contributing to the complexity and diversity of biological systems. In abnormal cells, it can also lead to deficiencies in protein function, potentially leading to pathogenesis of diseases. Detecting such events for single-gene genetic traits is relatively uncomplicated; however, the heterogeneity of populations with complex diseases presents an intricate challenge due to the presence of diverse causal events and undetermined subtypes. SPIT is the first statistical tool that quantifies the heterogeneity in transcript usage within a population and identifies predominant subgroups along with their distinctive sets of DTU events. We provide comprehensive assessments of SPIT's methodology in both single-gene and complex traits and report the results of applying SPIT to analyze brain samples from individuals with schizophrenia. Our analysis reveals previously unreported DTU events in six candidate genes.
Collapse
Affiliation(s)
- Beril Erdogdu
- Center for Computational Biology, Johns Hopkins University; Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering; Baltimore, MD, United States
| | - Ales Varabyou
- Center for Computational Biology, Johns Hopkins University; Baltimore, MD, United States
- Department of Computer Science, Johns Hopkins University; Baltimore, MD, United States
| | - Stephanie C Hicks
- Center for Computational Biology, Johns Hopkins University; Baltimore, MD, United States
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, MD, USA
| | - Steven L Salzberg
- Center for Computational Biology, Johns Hopkins University; Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering; Baltimore, MD, United States
- Department of Computer Science, Johns Hopkins University; Baltimore, MD, United States
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine; Baltimore, MD, United States
| | - Mihaela Pertea
- Center for Computational Biology, Johns Hopkins University; Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins School of Medicine and Whiting School of Engineering; Baltimore, MD, United States
- Department of Computer Science, Johns Hopkins University; Baltimore, MD, United States
- Department of Genetic Medicine, Johns Hopkins School of Medicine; Baltimore, MD, United States
| |
Collapse
|
13
|
Voraberger B, Mayr JA, Fratzl-Zelman N, Blouin S, Uday S, Kopajtich R, Koedam M, Hödlmayr H, Wortmann SB, Csillag B, Prokisch H, van der Eerden BCJ, El-Gazzar A, Högler W. Investigating the role of ASCC1 in the causation of bone fragility. Front Endocrinol (Lausanne) 2023; 14:1137573. [PMID: 37455927 PMCID: PMC10348481 DOI: 10.3389/fendo.2023.1137573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Bi-allelic variants in ASCC1 cause the ultrarare bone fragility disorder "spinal muscular atrophy with congenital bone fractures-2" (SMABF2). However, the mechanism by which ASCC1 dysfunction leads to this musculoskeletal condition and the nature of the associated bone defect are poorly understood. By exome sequencing, we identified a novel homozygous deletion in ASCC1 in a female infant. She was born with severe muscular hypotonia, inability to breathe and swallow, and virtual absence of spontaneous movements; showed progressive brain atrophy, gracile long bones, very slender ribs, and a femur fracture; and died from respiratory failure aged 3 months. A transiliac bone sample taken postmortem revealed a distinct microstructural bone phenotype with low trabecular bone volume, low bone remodeling, disordered collagen organization, and an abnormally high bone marrow adiposity. Proteomics, RNA sequencing, and qPCR in patient-derived skin fibroblasts confirmed that ASCC1 was hardly expressed on protein and RNA levels compared with healthy controls. Furthermore, we demonstrate that mutated ASCC1 is associated with a downregulation of RUNX2, the master regulator of osteoblastogenesis, and SERPINF1, which is involved in osteoblast and adipocyte differentiation. It also exerts an inhibitory effect on TGF-β/SMAD signaling, which is important for bone development. Additionally, knockdown of ASCC1 in human mesenchymal stromal cells (hMSCs) suppressed their differentiation capacity into osteoblasts while increasing their differentiation into adipocytes. This resulted in reduced mineralization and elevated formation of lipid droplets. These findings shed light onto the pathophysiologic mechanisms underlying SMABF2 and assign a new biological role to ASCC1 acting as an important pro-osteoblastogenic and anti-adipogenic regulator.
Collapse
Affiliation(s)
- Barbara Voraberger
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Johannes A. Mayr
- University Children’s Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Nadja Fratzl-Zelman
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Center Meidling, 1 Medical Department, Hanusch Hospital, Vienna, Austria
- Vienna Bone and Growth Center, Vienna, Austria
| | - Stéphane Blouin
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of OEGK and AUVA Trauma Center Meidling, 1 Medical Department, Hanusch Hospital, Vienna, Austria
- Vienna Bone and Growth Center, Vienna, Austria
| | - Suma Uday
- Department of Endocrinology and Diabetes, Birmingham Women’s and Children’s NHS Foundation Trust, Institute of Metabolism and Systems Research, University of Birmingham Edgbaston, Birmingham, United Kingdom
| | - Robert Kopajtich
- Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Marijke Koedam
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Helena Hödlmayr
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Saskia B. Wortmann
- University Children’s Hospital Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
- Amalia Children’s Hospital, Radboudumc, Nijmegen, Netherlands
| | - Bernhard Csillag
- Department of Neonatology, Kepler University Hospital, Linz, Austria
| | - Holger Prokisch
- Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Bram C. J. van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ahmed El-Gazzar
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
14
|
Guadagnolo D, Mastromoro G, Hashemian NK, Orlando V, Lepri FR, D'Alberti E, Giancotti A, Novelli A, Pizzuti A. Fetal first-trimester cystic hygroma as the prenatal presenting feature of ASCC1-related Spinal Muscular Atrophy with Bone Fractures 2. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2023:S1701-2163(23)00373-0. [PMID: 37201580 DOI: 10.1016/j.jogc.2023.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023]
Affiliation(s)
- Daniele Guadagnolo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Gioia Mastromoro
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Valeria Orlando
- Laboratory of Medical Genetics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Elena D'Alberti
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella Giancotti
- Department of Maternal, Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Antonio Pizzuti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
15
|
Jia J, Hilal T, Bohnsack KE, Chernev A, Tsao N, Bethmann J, Arumugam A, Parmely L, Holton N, Loll B, Mosammaparast N, Bohnsack MT, Urlaub H, Wahl MC. Extended DNA threading through a dual-engine motor module of the activating signal co-integrator 1 complex. Nat Commun 2023; 14:1886. [PMID: 37019967 PMCID: PMC10076317 DOI: 10.1038/s41467-023-37528-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
Activating signal co-integrator 1 complex (ASCC) subunit 3 (ASCC3) supports diverse genome maintenance and gene expression processes, and contains tandem Ski2-like NTPase/helicase cassettes crucial for these functions. Presently, the molecular mechanisms underlying ASCC3 helicase activity and regulation remain unresolved. We present cryogenic electron microscopy, DNA-protein cross-linking/mass spectrometry as well as in vitro and cellular functional analyses of the ASCC3-TRIP4 sub-module of ASCC. Unlike the related spliceosomal SNRNP200 RNA helicase, ASCC3 can thread substrates through both helicase cassettes. TRIP4 docks on ASCC3 via a zinc finger domain and stimulates the helicase by positioning an ASC-1 homology domain next to the C-terminal helicase cassette of ASCC3, likely supporting substrate engagement and assisting the DNA exit. TRIP4 binds ASCC3 mutually exclusively with the DNA/RNA dealkylase, ALKBH3, directing ASCC3 for specific processes. Our findings define ASCC3-TRIP4 as a tunable motor module of ASCC that encompasses two cooperating NTPase/helicase units functionally expanded by TRIP4.
Collapse
Affiliation(s)
- Junqiao Jia
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustr. 6, D-14195, Berlin, Germany
- Harvard Medical School, Department of Cell Biology, 240 Longwood Avenue, Boston, MA, 02115, USA
| | - Tarek Hilal
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustr. 6, D-14195, Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Research Center of Electron Microscopy, Fabeckstr. 36a, D-14195, Berlin, Germany
| | - Katherine E Bohnsack
- Universitätsmedizin Göttingen, Department of Molecular Biology, Humboldallee 23, D-37073, Göttingen, Germany
| | - Aleksandar Chernev
- Max-Planck-Institut für Multidisziplinäre Naturwissenschaften, Bioanalytical Mass Spectrometry, Am Fassberg 11, D-37077, Göttingen, Germany
| | - Ning Tsao
- Washington University School of Medicine, Department of Pathology & Immunology and Center for Genome Integrity, 660 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Juliane Bethmann
- Max-Planck-Institut für Multidisziplinäre Naturwissenschaften, Bioanalytical Mass Spectrometry, Am Fassberg 11, D-37077, Göttingen, Germany
- Universitätsmedizin Göttingen, Institut für Klinische Chemie, Bioanalytik, Robert-Koch-Straße 40, D-35075, Göttingen, Germany
| | - Aruna Arumugam
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustr. 6, D-14195, Berlin, Germany
| | - Lane Parmely
- Washington University School of Medicine, Department of Pathology & Immunology and Center for Genome Integrity, 660 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Nicole Holton
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustr. 6, D-14195, Berlin, Germany
| | - Bernhard Loll
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustr. 6, D-14195, Berlin, Germany
| | - Nima Mosammaparast
- Washington University School of Medicine, Department of Pathology & Immunology and Center for Genome Integrity, 660 S. Euclid Ave, St. Louis, MO, 63110, USA
| | - Markus T Bohnsack
- Universitätsmedizin Göttingen, Department of Molecular Biology, Humboldallee 23, D-37073, Göttingen, Germany
- Georg-August-Universität, Göttingen Center for Molecular Biosciences, Justus-von-Liebig-Weg 11, D-37077, Göttingen, Germany
- Max-Planck-Institut für Multidisziplinäre Naturwissenschaften, Am Fassberg 11, D-37077, Göttingen, Germany
| | - Henning Urlaub
- Max-Planck-Institut für Multidisziplinäre Naturwissenschaften, Bioanalytical Mass Spectrometry, Am Fassberg 11, D-37077, Göttingen, Germany
- Universitätsmedizin Göttingen, Institut für Klinische Chemie, Bioanalytik, Robert-Koch-Straße 40, D-35075, Göttingen, Germany
| | - Markus C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Takustr. 6, D-14195, Berlin, Germany.
- Helmholtz-Zentrum Berlin für Materialien und Energie, Macromolecular Crystallography, Albert-Einstein-Str. 15, D-12489, Berlin, Germany.
| |
Collapse
|
16
|
Cao Z, Wang H, Chen J, Zhang Y, Mo Q, Zhang P, Wang M, Liu H, Bao X, Sun Y, Zhang W, Yao Q. Silk-based hydrogel incorporated with metal-organic framework nanozymes for enhanced osteochondral regeneration. Bioact Mater 2023; 20:221-242. [PMID: 35702612 PMCID: PMC9163388 DOI: 10.1016/j.bioactmat.2022.05.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Osteochondral defects (OCD) cannot be efficiently repaired due to the unique physical architecture and the pathological microenvironment including enhanced oxidative stress and inflammation. Conventional strategies, such as the control of implant microstructure or the introduction of growth factors, have limited functions failing to manage these complex environments. Here we developed a multifunctional silk-based hydrogel incorporated with metal-organic framework nanozymes (CuTA@SF) to provide a suitable microenvironment for enhanced OCD regeneration. The incorporation of CuTA nanozymes endowed the SF hydrogel with a uniform microstructure and elevated hydrophilicity. In vitro cultivation of mesenchymal stem cells (MSCs) and chondrocytes showed that CuTA@SF hydrogel accelerated cell proliferation and enhanced cell viability, as well as had antioxidant and antibacterial properties. Under the inflammatory environment with the stimulation of IL-1β, CuTA@SF hydrogel still possessed the potential to promote MSC osteogenesis and deposition of cartilage-specific extracellular matrix (ECM). The proteomics analysis further confirmed that CuTA@SF hydrogel promoted cell proliferation and ECM synthesis. In the full-thickness OCD model of rabbit, CuTA@SF hydrogel displayed successfully in situ OCD regeneration, as evidenced by micro-CT, histology (HE, S/O, and toluidine blue staining) and immunohistochemistry (Col I and aggrecan immunostaining). Therefore, CuTA@SF hydrogel is a promising biomaterial targeted at the regeneration of OCD.
Collapse
Affiliation(s)
- Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Hongmei Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Pharmaceutical Sciences, Binzhou Medical University, 264003, Yantai, Shandong, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Yanan Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xueyang Bao
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| |
Collapse
|
17
|
Bouman K, Dittrich AT, Groothuis JT, van Engelen BG, Janssen MC, Voermans NC, Draaisma JM, Erasmus CE. Bone Quality in Patients with a Congenital Myopathy: A Scoping Review. J Neuromuscul Dis 2023; 10:1-13. [PMID: 36314217 PMCID: PMC9881028 DOI: 10.3233/jnd-221543] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Congenital myopathies are rare neuromuscular disorders presenting with a wide spectrum of clinical features, including long bone fractures (LBFs) that negatively influence functional prognosis, quality of life and survival. Systematic research on bone quality in these patients is lacking. OBJECTIVE This scoping review aims to summarize all evidence on bone quality and to deduce recommendations for bone quality management in congenital myopathies. METHODS Five electronic databases (Pubmed, Embase, Cochrane, Web of Science, CINAHL) were searched. All studies on bone quality in congenital myopathies were included. Decreased bone quality was defined as low bone mineral density and/or (fragility) LBFs. Study selection and data extraction were performed by three independent reviewers. RESULTS We included 244 single cases (mean: 4.1±7.6 years; median: 0 years) diagnosed with a congenital myopathy from 35 articles. Bone quality was decreased in 93 patients (37%) (mean: 2.6±6.8 years; median: 0 years). Low bone mineral density was reported in 11 patients (4.5%) (mean: 10.9±9.7; median: 11 years). Congenital LBFs were reported in 64 patients (26%). (Fragility) LBFs later at life were described in 24 patients (9.8%) (mean: 14.9±11.0; median: 14 years). Four cases (1.6%) were reported to receive vitamin D and/or calcium supplementation or diphosphonate administration. CONCLUSION LBFs are thus frequently reported in congenital myopathies. We therefore recommend optimal bone quality management through bone mineral density assessment, vitamin D and calcium suppletion, and referral to internal medicine or pediatrics for consideration of additional therapies in order to prevent complications of low bone mineral density.
Collapse
Affiliation(s)
- Karlijn Bouman
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anne T.M. Dittrich
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan T. Groothuis
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Baziel G.M. van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mirian C.H. Janssen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nicol C. Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jos M.T. Draaisma
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Corrie E. Erasmus
- Department of Pediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Yu S, Zhao H, Meng H, Shi S, Cao S, Bian T, Ruan C. CSRP1 Promotes Colon Adenocarcinoma Growth and Serves as an Independent Risk Biomarker for Worse Prognosis. Genet Res (Camb) 2023; 2023:8586507. [PMID: 37113556 PMCID: PMC10129416 DOI: 10.1155/2023/8586507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Background Cysteine and Glycine Rich Protein 1 (CSRP1) belongs to the cysteine-rich protein family, which contains a unique double-zinc finger motif and is important for development and cellular differentiation. Abnormal expression of CSRP1 was reported within several malignancies such as prostate cancer and acute myeloid leukemia. Here, we explored function of CSRP1 within colon adenocarcinoma (COAD) for the first time. Methods The mRNA levels of CSRP1 in COADs were obtained from TCGA datasets. CSRP1 protein expressions in COADs were tested via immunohistochemistry staining. Patients' prognosis was evaluated using both univariate analysis and multivariate analysis. Two human COAD originated cancer cell lines, Caco-2, and HT-29, were used for cellular experiments including shRNA knockdown, proliferation assay, and migration assay. In vivo model was established using nude mice xenografts to further validate the role of CSRP1 in COAD progression. Results The mRNA levels of CSRP1 are elevated in COAD specimens from patients with more advanced tumor stages and higher Carcinoembryonic Antigen (CEA) levels. In addition, higher CSRP1 mRNA level indicates worse COAD prognosis. Consistently, higher CSRP1 protein expression is correlated with worse overall survival according to both univariate and multivariate analysis, indicating that CSRP1 is a new COAD prognostic factor. Furthermore, COAD cells transfected with CSRP1-shRNAs exhibit attenuated proliferation and migration capacities. Finally, growth of xenografts originated from CSRP1-knockdown cells is inhibited comparing to the control ones. Conclusions Expression of CSRP1 is positively correlated with COAD progression, which can promote tumor growth and migration. Higher CSRP1 can is a novel independent prognostic factor of COAD.
Collapse
Affiliation(s)
- Senlong Yu
- Department of Gastrointestinal Surgery, Zhuji People's Hospital of Zhejiang Province (Zhuji Affiliated Hospital of Shaoxing University), Zhuji 311800, China
| | - Haifeng Zhao
- Department of General Surgery, Zhuji Chinese Traditional Medical Hospital, Zhuji 311800, China
| | - Hongjie Meng
- Department of Gastrointestinal Surgery, Zhuji People's Hospital of Zhejiang Province (Zhuji Affiliated Hospital of Shaoxing University), Zhuji 311800, China
| | - Shengguang Shi
- Department of Gastrointestinal Surgery, Zhuji People's Hospital of Zhejiang Province (Zhuji Affiliated Hospital of Shaoxing University), Zhuji 311800, China
| | - Shenghui Cao
- Department of General Surgery, Zhuji Chinese Traditional Medical Hospital, Zhuji 311800, China
| | - Tianhua Bian
- Department of General Surgery, Zhuji Chinese Traditional Medical Hospital, Zhuji 311800, China
| | - Canping Ruan
- Colorectal Surgery Department, Changzheng Hospital, Shanghai 200003, China
| |
Collapse
|
19
|
Sharova M, Guseva D, Kurenkov A, Novoselova O, Murtazina A, Skoblov M. Congenital myopathy as a new phenotype caused by two undescribed variants in ASCC1 gene. Am J Med Genet A 2022; 188:3100-3105. [PMID: 35838082 DOI: 10.1002/ajmg.a.62898] [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: 07/29/2021] [Revised: 05/19/2022] [Accepted: 06/02/2022] [Indexed: 01/31/2023]
Abstract
We present a patient with congenital myopathy and an inborn epiphysiolysis of the ulna. Whole-exome sequencing analysis revealed two novel mutations in Activation Signal Cointegrator Complex 1 (ASCC1) gene in a compound heterozygous state-a splicing variant c.395-2A>G and a deletion of the first two coding exons. Homozygous and compound heterozygous LoF variants in ASCC1 gene lead to a severe phenotype of spinal muscular atrophy with congenital bone fractures 2 (SMABF2). All patients described to date presented with a severe muscular hypotony, inborn fractures, and passed away shortly after birth while our proband had moderate hypotony, no fractures, but epiphysiolysis and he was 3.5 years old at the time of examination. To explain the phenotype of our patient, we performed an RNA analysis of all family members. We discovered that the c.395-2A>G variant results in two aberrant mRNA isoforms. We also validated the deletion of two exons in ASCC1 gene that lead to the increased expression of this truncated transcript by 1.8 times. To investigate the possible impact of this deletion on the phenotype we predicted a new Kozak sequence in exon 4 that could lead to the formation of a truncated protein with shortened KH domain and a full RNA ligase-like domain. We suggest that this unexpectedly different phenotype of the proband with ASCC1-related disorder could be explained by the presence of the truncated protein with an increased expression.
Collapse
Affiliation(s)
| | - Darya Guseva
- Research Centre for Medical Genetics, Moscow, Russia
| | - Alexey Kurenkov
- National Medical Research Center for Children's Health, the Russian Federation Ministry of Healthcare, Moscow, Russia
| | - Olga Novoselova
- Genomed Ltd., Genetic Diagnostic Lab, Moscow, Russia.,Filatov N.F. Children's City Hospital 103001, Moscow, Russia
| | | | | |
Collapse
|
20
|
Marais A, Bertoli-Avella AM, Beetz C, Altunoglu U, Alhashem A, Mohamed S, Alghamdi A, Willems P, Tsoutsou E, Fryssira H, Pons R, Almarzooq R, Karatoprak EY, Ayaz A, Ünverengil G, Calvo M, Yüksel Z, Bauer P. Further clinical and genetic evidence of ASC-1 complex dysfunction in congenital neuromuscular disease. Eur J Med Genet 2022; 65:104537. [PMID: 35690317 DOI: 10.1016/j.ejmg.2022.104537] [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: 05/19/2021] [Revised: 05/21/2022] [Accepted: 06/06/2022] [Indexed: 11/03/2022]
Abstract
Transcriptional coregulators modulate the efficiency of transcription factors. Bi-allelic variants in TRIP4 and ASCC1, two genes that encode members of the tetrameric coregulator ASC-1, have recently been associated with congenital bone fractures, hypotonia, and muscular dystrophy in a total of 22 unrelated families. Upon exome sequencing and data repository mining, we identified six new patients with pathogenic homozygous variants in either TRIP4 (n = 4, two novel variants) or ASCC1 (n = 2, one novel variant). The associated clinical findings confirm and extend previous descriptions. Considering all patients reported to date, we provide supporting evidence suggesting that ASCC1-related disease has a more severe phenotype compared to TRIP4-related disorder regarding higher incidence of perinatal bone fractures and shorter survival.
Collapse
Affiliation(s)
| | | | | | - Umut Altunoglu
- Department of Medical Genetics, Koç University, School of Medicine, 34450, Istanbul, Turkey
| | - Amal Alhashem
- Division of Genetics and Metabolic Medicine, Department of Pediatrics, Prince Sultan Military Medical City Riyadh, Saudi Arabia; Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Sarar Mohamed
- Division of Genetics and Metabolic Medicine, Department of Pediatrics, Prince Sultan Military Medical City Riyadh, Saudi Arabia; Prince Abdullah Bin Khaled Coeliac Disease Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Pediarics, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Abdulaziz Alghamdi
- Pediatrics Department, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | | | - Eirini Tsoutsou
- Medical Genetics Department, Choremio Research Laboratory, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, Greece
| | - Helena Fryssira
- Medical Genetics Department, Choremio Research Laboratory, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, Greece
| | - Roser Pons
- Medical Genetics Department, Choremio Research Laboratory, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, Greece
| | - Reem Almarzooq
- Pediatric Department, Salmaniya Medical Complex, Manama, Bahrain
| | - Elif Yüksel Karatoprak
- Departments of Pediatric Neurology, İstanbul Medeniyet University Faculty of Medicine, Göztepe Training and Research Hospital, İstanbul, Turkey
| | - Akif Ayaz
- Istanbul Medipol University Faculty of Medicine, Department of Medical Genetics, Istanbul, Turkey
| | - Gökçen Ünverengil
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | | | | | | |
Collapse
|
21
|
Bradinova I, Andonova S, Vazharova R, Tomova S, Balabanski L, Savov A. Spinal muscular atrophy with congenital bone fractures 2 caused by a rare loss-of-function ASCC1 gene mutation in two Bulgarian Roma patients. Clin Genet 2022; 102:78-79. [PMID: 35338657 DOI: 10.1111/cge.14130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/08/2022] [Accepted: 03/16/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Irena Bradinova
- Department of Obstetrics and Gynecology, National Genetic Laboratory, Medical University Sofia, Sofia, Bulgaria
| | - Silvia Andonova
- Department of Obstetrics and Gynecology, National Genetic Laboratory, Medical University Sofia, Sofia, Bulgaria
| | - Radoslava Vazharova
- Faculty of Medicine, Department of Biology, Medical Genetics and Microbiology, SU "St. Kl. Ohridski", Sofia, Bulgaria
| | - Stiliyana Tomova
- Department of Neonatology, University Hospital MBAL Bourgas, Burgas, Bulgaria
| | | | - Alexey Savov
- Department of Obstetrics and Gynecology, National Genetic Laboratory, Medical University Sofia, Sofia, Bulgaria
| |
Collapse
|
22
|
ASC1 complex related conditions: Two novel paediatric patients with TRIP4 pathogenic variants and review of literature. Eur J Med Genet 2022; 65:104469. [PMID: 35276412 DOI: 10.1016/j.ejmg.2022.104469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/11/2022] [Accepted: 02/25/2022] [Indexed: 11/22/2022]
Abstract
Pathogenic variants in the genes encoding for the ASC1 complex were recently reported in patients with congenital fractures, joint contractures, neonatal hypotonia and respiratory distress. Here we report two male children with biallelic TRIP4 pathogenic loss of function variants. The first child presented with foetal bradykinesia, neonatal respiratory distress, central and peripheral hypotonia, constipation, hyperlaxity, left uretero-hydronephrosis and post-obstructive kidney dysplasia. The second had severe central and peripheral neonatal hypotonia, feeding difficulties, kyphosis, developmental delay and hyperlaxity. Detailed review of all reported cases with ASCC1 (12 patients) and TRIP4 (18 patients) variants highlights striking genotype-phenotype correlations. This is the fourth report of patients with TRIP4 variants and the first description of post-obstructive kidney dysplasia in this condition.
Collapse
|
23
|
Mukherjee S, Park JP, Yun JW. Carboxylesterase3 (Ces3) Interacts with Bone Morphogenetic Protein 11 and Promotes Differentiation of Osteoblasts via Smad1/5/9 Pathway. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-021-0133-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
24
|
Li W, Hu S, Tian C, Wan X, Yu W, Guo P, Zhao F, Hua C, Lu X, Xue G, Han S, Guo W, Wang D, Deng W. TRIP4 transcriptionally activates DDIT4 and subsequent mTOR signaling to promote glioma progression. Free Radic Biol Med 2021; 177:31-47. [PMID: 34648907 DOI: 10.1016/j.freeradbiomed.2021.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/25/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022]
Abstract
In spite of significant advances in the understanding of glioma biology and pathology, survival remains poor. Therefore, it is still of great significance to further explore the key factors involved in tumorigenesis and development in glioma and find potential new therapeutic targets. Here, we show that thyroid hormone receptor interactor 4 (TRIP4) is highly expressed in glioma cells and tissues. Patients of glioma with high expression of TRIP4 possess poor overall survival. Knockdown of TRIP4 inhibited tumor cell proliferation, metastasis, and apoptosis suppression, whereas overexpression of TRIP4 displays the opposite effects. Further research showed that TRIP4 promoted glioma progression through regulating DDIT4 expression and subsequent activation of mTOR signaling. DDIT4 overexpression restored the inhibition of tumor growth by TRIP4 knockdown in vitro and in vivo. Consistently, mTOR activity inhibition reversed TRIP4 overexpression-mediated tumor promotion in vitro and in vivo. Moreover, molecular mechanism exploration demonstrates that TRIP4 functions as a specific transcriptional activator to anchor at the promoter region of DDIT4 gene (-196 to -11) to regulate its transcription and such regulation was affected by HIF1α. Clinically, TRIP4 expression is positively correlated with DDIT4 expression in glioma samples based on tissue microarray analysis and both of their high expression predicts the malignancy of the disease. Altogether, our findings identify TRIP4 as a critical promoter of glioma progression by targeting DDIT4 and mTOR signaling successively and suggest that TRIP4-DDIT4 axis has potential to be a novel therapeutic target in glioma treatment.
Collapse
Affiliation(s)
- Wenyang Li
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Sheng Hu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Chunfang Tian
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Xinyu Wan
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Ping Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Feng Zhao
- Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Chunyu Hua
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Xiaona Lu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Guoqing Xue
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Shilong Han
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Wei Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China.
| | - Dong Wang
- Department of Hepatobiliary Surgery, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| |
Collapse
|
25
|
Töpf A, Pyle A, Griffin H, Matalonga L, Schon K, Sickmann A, Schara-Schmidt U, Hentschel A, Chinnery PF, Kölbel H, Roos A, Horvath R. Exome reanalysis and proteomic profiling identified TRIP4 as a novel cause of cerebellar hypoplasia and spinal muscular atrophy (PCH1). Eur J Hum Genet 2021; 29:1348-1353. [PMID: 34075209 PMCID: PMC8440675 DOI: 10.1038/s41431-021-00851-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/12/2021] [Accepted: 02/26/2021] [Indexed: 01/26/2023] Open
Abstract
TRIP4 is one of the subunits of the transcriptional coregulator ASC-1, a ribonucleoprotein complex that participates in transcriptional coactivation and RNA processing events. Recessive variants in the TRIP4 gene have been associated with spinal muscular atrophy with bone fractures as well as a severe form of congenital muscular dystrophy. Here we present the diagnostic journey of a patient with cerebellar hypoplasia and spinal muscular atrophy (PCH1) and congenital bone fractures. Initial exome sequencing analysis revealed no candidate variants. Reanalysis of the exome data by inclusion in the Solve-RD project resulted in the identification of a homozygous stop-gain variant in the TRIP4 gene, previously reported as disease-causing. This highlights the importance of analysis reiteration and improved and updated bioinformatic pipelines. Proteomic profile of the patient's fibroblasts showed altered RNA-processing and impaired exosome activity supporting the pathogenicity of the detected variant. In addition, we identified a novel genetic form of PCH1, further strengthening the link of this characteristic phenotype with altered RNA metabolism.
Collapse
Affiliation(s)
- Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Angela Pyle
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Helen Griffin
- Primary Immunodeficiency Group, Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Leslie Matalonga
- CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Katherine Schon
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- MRC Mitochondrial Biology Unit, Cambridge Biomedical Campus, Cambridge, UK
| | - Albert Sickmann
- Department of Bioanalytics, Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
- Medizinische Proteom-Center (MPC), Medizinische Fakultät, Ruhr-Universität Bochum, Bochum, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children's Hospital University of Essen, Essen, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- MRC Mitochondrial Biology Unit, Cambridge Biomedical Campus, Cambridge, UK
| | - Heike Kölbel
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children's Hospital University of Essen, Essen, Germany
| | - Andreas Roos
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children's Hospital University of Essen, Essen, Germany.
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| |
Collapse
|
26
|
Juntas Morales R, Perrin A, Solé G, Lacourt D, Pegeot H, Walther-Louvier U, Cintas P, Cances C, Espil C, Theze C, Zenagui R, Yauy K, Cosset E, Renard D, Rigau V, Maues de Paula A, Uro-Coste E, Arne-Bes MC, Martin Négrier ML, Leboucq N, Acket B, Malfatti E, Biancalana V, Metay C, Richard P, Rendu J, Rivier F, Koenig M, Cossée M. An Integrated Clinical-Biological Approach to Identify Interindividual Variability and Atypical Phenotype-Genotype Correlations in Myopathies: Experience on A Cohort of 156 Families. Genes (Basel) 2021; 12:genes12081199. [PMID: 34440373 PMCID: PMC8392536 DOI: 10.3390/genes12081199] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 01/17/2023] Open
Abstract
Diagnosis of myopathies is challenged by the high genetic heterogeneity and clinical overlap of the various etiologies. We previously reported a Next-Generation Sequencing strategy to identify genetic etiology in patients with undiagnosed Limb-Girdle Muscular Dystrophies, Congenital Myopathies, Congenital Muscular Dystrophies, Distal Myopathies, Myofibrillar Myopathies, and hyperCKemia or effort intolerance, using a large gene panel including genes classically associated with other entry diagnostic categories. In this study, we report the comprehensive clinical-biological strategy used to interpret NGS data in a cohort of 156 pediatric and adult patients, that included Copy Number Variants search, variants filtering and interpretation according to ACMG guidelines, segregation studies, deep phenotyping of patients and relatives, transcripts and protein studies, and multidisciplinary meetings. Genetic etiology was identified in 74 patients, a diagnostic yield (47.4%) similar to previous studies. We identified 18 patients (10%) with causative variants in different genes (ACTA1, RYR1, NEB, TTN, TRIP4, CACNA1S, FLNC, TNNT1, and PAPBN1) that resulted in milder and/or atypical phenotypes, with high intrafamilial variability in some cases. Mild phenotypes could mostly be explained by a less deleterious effect of variants on the protein. Detection of inter-individual variability and atypical phenotype-genotype associations is essential for precision medicine, patient care, and to progress in the understanding of the molecular mechanisms of myopathies.
Collapse
Affiliation(s)
- Raul Juntas Morales
- Explorations Neurologiques et Centre SLA, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), Centre Hospitalier Universitaire de Montpellier, 34295 Montpellier, France;
- Équipe Accueil EA7402, Institut Universitaire de Recherche Clinique (IURC), Université de Montpellier, 34093 Montpellier, France;
| | - Aurélien Perrin
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | - Guilhem Solé
- Service de Neurologie, Centre Hospitalier Universitaire de Bordeaux, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 33000 Bordeaux, France;
| | - Delphine Lacourt
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Henri Pegeot
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Ulrike Walther-Louvier
- Service de Neuropédiatrie, Centre Hospitalier Universitaire de Montpellier, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France; (U.W.-L.); (F.R.)
| | - Pascal Cintas
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France; (P.C.); (M.-C.A.-B.); (B.A.)
| | - Claude Cances
- Service de Neuropédiatrie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France;
| | - Caroline Espil
- Service de Neuropédiatrie, Centre Hospitalier de Bordeaux, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 33000 Bordeaux, France;
| | - Corinne Theze
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Reda Zenagui
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Kevin Yauy
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Elodie Cosset
- Équipe Accueil EA7402, Institut Universitaire de Recherche Clinique (IURC), Université de Montpellier, 34093 Montpellier, France;
| | - Dimitri Renard
- Service de Neurologie, Centre Hospitalier Universitaire de Nîmes, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 30029 Nîmes, France;
| | - Valerie Rigau
- Service de Pathologie, Centre Hospitalier Universitaire de Montpellier, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France;
| | - Andre Maues de Paula
- Service de Pathologie, Centre Hospitalier Universitaire de Marseille, Centre de Référence des Maladies Neuromusculaires PACA-Réunion-Rhône Alpes, 13005 Marseille, France;
| | - Emmanuelle Uro-Coste
- Service de Pathologie, Centre Hospitalier Universitaire de Toulouse, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31300 Toulouse, France;
| | - Marie-Christine Arne-Bes
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France; (P.C.); (M.-C.A.-B.); (B.A.)
| | - Marie-Laure Martin Négrier
- CHU de Bordeaux, Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, 33076 Bordeaux, France;
| | - Nicolas Leboucq
- Service de Neuroradiologie, Centre Hospitalier de Montpellier, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France;
| | - Blandine Acket
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France; (P.C.); (M.-C.A.-B.); (B.A.)
| | - Edoardo Malfatti
- Service Neurologie Médicale, Centre de Référence Maladies Neuromusculaires Nord-Est-Ile-de-France, CHU Raymond-Poincaré, 92380 Garches, France;
- U1179 UVSQ-INSERM Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, UFR des Sciences de la Santé Simone Veil, Université Versailles-Saint-Quentin-en-Yvelines, 78180 Versailles, France
| | - Valérie Biancalana
- Laboratoire de Diagnostic Génétique, Université de Strasbourg, 67084 Strasbourg, France;
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, Université de Strasbourg, 67404 Illkirch, France
| | - Corinne Metay
- Unité Fonctionnelle de Cardiogénétique et Myogénétique, Centre de Génétique, Hôpitaux Universitaire Pitié Salpêtrière–Charles Foix, 75651 Paris, France; (C.M.); (P.R.)
| | - Pascale Richard
- Unité Fonctionnelle de Cardiogénétique et Myogénétique, Centre de Génétique, Hôpitaux Universitaire Pitié Salpêtrière–Charles Foix, 75651 Paris, France; (C.M.); (P.R.)
| | - John Rendu
- CHU Grenoble, Université de Grenoble Alpes, Inserm, U1216, GIN, 38706 Saint-Martin-d’Hères, France;
- Unité Médicale de Génétique Moléculaire, Centre Hospitalier, Universitaire Grenoble Alpes, 38043 Saint-Martin-d’Hères, France
| | - François Rivier
- Service de Neuropédiatrie, Centre Hospitalier Universitaire de Montpellier, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France; (U.W.-L.); (F.R.)
| | - Michel Koenig
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | - Mireille Cossée
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34093 Montpellier, France
- Correspondence:
| |
Collapse
|
27
|
Correa ARE, Naini K, Mishra P, Dadhwal V, Agarwal R, Shukla R, Kabra M, Gupta N. Utility of fetal whole exome sequencing in the etiological evaluation and outcome of nonimmune hydrops fetalis. Prenat Diagn 2021; 41:1414-1424. [PMID: 34302381 DOI: 10.1002/pd.6022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/21/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Nonimmune hydrops fetalis (NIHF) has varied etiology. We assessed the etiological spectrum and evaluated the utility of fetal whole exome sequencing (fWES) for the diagnosis of NIHF. METHODS In this prospective cohort study, we evaluated antenatally diagnosed fetuses with NIHF between July 2018 and December 2019 according to the routine diagnostic algorithm. Fetuses that remained undiagnosed after routine NIHF workup were subjected to fetal chromosomal microarray and/or WES. Pregnancies were followed up for clinical outcomes. RESULTS Of the 45 fetuses, consanguinity and recurrent hydrops fetalis were observed in 13.3% (6/45) and 28.8% (13/45), respectively. Overall, an etiological diagnosis was possible in 75.5% (34/45) of fetuses, while the cause remained unknown in 24.4% (11/45). A genetic etiology was identified in 46.6% (21/45): aneuploidy and monogenic disorders in 28.8% (13/45) and 17.8% (8/45), respectively. fWES on 19 fetuses detected disease-causing variants in 42.1% (8/19). Nine novel variants were detected in RAPSN, ASCC1, NEB, PKD1L1, GUSB, and PIEZO1. Only 8.8% (4/45) of the cohort survived without morbidity. CONCLUSIONS This study describes the etiological spectrum and the disease-causing variants in an Indian cohort of hydropic fetuses.
Collapse
Affiliation(s)
- Alec Reginald Errol Correa
- Department of Pediatrics, Division of Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Kamal Naini
- Department of Pediatrics, Division of Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Pallavi Mishra
- Department of Pediatrics, Division of Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Vatsla Dadhwal
- Department of Obstetrics & Gynecology, All India Institute of Medical Sciences, New Delhi, India
| | - Ramesh Agarwal
- Department of Pediatrics, Division of Neonatology, All India Institute of Medical Sciences, New Delhi, India
| | - Rashmi Shukla
- Department of Pediatrics, Division of Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Madhulika Kabra
- Department of Pediatrics, Division of Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Neerja Gupta
- Department of Pediatrics, Division of Genetics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
28
|
Wang S, Wu P, Wang K, Ji X, Chen D, Jiang A, Liu Y, Xiao W, Jiang Y, Zhu L, Xu X, Li M, Li X, Tang G. Transcriptome Analysis Reveals Key Genes and Pathways Associated with Mummify Piglets. Genome 2021; 64:1029-1040. [PMID: 34139142 DOI: 10.1139/gen-2021-0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
China is the country with the largest pork consumption in the world. However, the incidence of high mummify piglets (3-5%) is one of the important factors that cause the slow improvement of pig reproductive capacity, and the genetic mechanism is still unclear. This study aimed to identify candidate genes related to high mummify piglets. RNA-seq technology was used to comparative transcriptome profiling of blood from high piglets mummified and healthy sow at different stages of pregnancy (35d, 56d, 77d and 98d). A total of 137 to 420 DEGs were detected in each stage. Seven differentially expressed genes were significantly differentially expressed at various stages. IL-9R, TLR8, ABLIM3, FSH-α, ASCC1, PRKCZ, and GCK may play an important role in course of mummify piglets. The differential genes we identified between the groups were mainly enriched in immune and inflammation regulation, and others were mainly enriched in reproduction. Considering the function of candidate genes, IL-9R and TLR8 were suggested as the most promising candidate genes involved in mummify piglet traits. We speculate that during pregnancy, it may be the combined effects of the above-mentioned inflammation, immune response, and reproduction-related signal pathways that affect the occurrence of mummifying piglets, and further affect pig reproduction.
Collapse
Affiliation(s)
- Shujie Wang
- Sichuan Agricultural University, 12529, Chengdu, Sichuan, China;
| | - Pingxian Wu
- Sichuan Agricultural University, 12529, Chengdu, Sichuan, China;
| | - Kai Wang
- Sichuan Agricultural University, 12529, Chengdu, Sichuan, China;
| | - Xiang Ji
- Sichuan Agricultural University, 12529, Chengdu, Sichuan, China;
| | - Dong Chen
- Sichuan Agricultural University, 12529, Chengdu, Sichuan, China;
| | - Anan Jiang
- Sichuan Agricultural University - Chengdu Campus, 506176, Chengdu, Sichuan, China;
| | - Yihui Liu
- Sichuan Animal Husbandry Station, Chengdu, Sichuan, China;
| | - Weihang Xiao
- Sichuan Agricultural University - Chengdu Campus, 506176, Chengdu, Sichuan, China;
| | - Yanzhi Jiang
- College of Life Science, Sichuan Agricultural University, Ya'an, China;
| | - Li Zhu
- Sichuan Agricultural University - Chengdu Campus, 506176, Chengdu, Sichuan, China;
| | - Xu Xu
- Sichuan Provincial Animal Husbandry and Food Bureau, 177358, Chengdu, Sichuan, China;
| | - Mingzhou Li
- Sichuan Agricultural University, 12529, Chengdu, Sichuan, China;
| | - Xuewei Li
- Sichuan Agricultural University - Chengdu Campus, 506176, Chengdu, Sichuan, China;
| | - Guoqing Tang
- Sichuan Agricultural University - Chengdu Campus, 506176, Chengdu, Sichuan, China;
| |
Collapse
|
29
|
Meunier J, Villar-Quiles RN, Duband-Goulet I, Ferreiro A. Inherited Defects of the ASC-1 Complex in Congenital Neuromuscular Diseases. Int J Mol Sci 2021; 22:ijms22116039. [PMID: 34204919 PMCID: PMC8199739 DOI: 10.3390/ijms22116039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Defects in transcriptional and cell cycle regulation have emerged as novel pathophysiological mechanisms in congenital neuromuscular disease with the recent identification of mutations in the TRIP4 and ASCC1 genes, encoding, respectively, ASC-1 and ASCC1, two subunits of the ASC-1 (Activating Signal Cointegrator-1) complex. This complex is a poorly known transcriptional coregulator involved in transcriptional, post-transcriptional or translational activities. Inherited defects in components of the ASC-1 complex have been associated with several autosomal recessive phenotypes, including severe and mild forms of striated muscle disease (congenital myopathy with or without myocardial involvement), but also cases diagnosed of motor neuron disease (spinal muscular atrophy). Additionally, antenatal bone fractures were present in the reported patients with ASCC1 mutations. Functional studies revealed that the ASC-1 subunit is a novel regulator of cell cycle, proliferation and growth in muscle and non-muscular cells. In this review, we summarize and discuss the available data on the clinical and histopathological phenotypes associated with inherited defects of the ASC-1 complex proteins, the known genotype–phenotype correlations, the ASC-1 pathophysiological role, the puzzling question of motoneuron versus primary muscle involvement and potential future research avenues, illustrating the study of rare monogenic disorders as an interesting model paradigm to understand major physiological processes.
Collapse
Affiliation(s)
- Justine Meunier
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, 75013 Paris, France; (J.M.); (R.-N.V.-Q.)
| | - Rocio-Nur Villar-Quiles
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, 75013 Paris, France; (J.M.); (R.-N.V.-Q.)
- Reference Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, APHP, Institute of Myology, 75013 Paris, France
| | - Isabelle Duband-Goulet
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, 75013 Paris, France; (J.M.); (R.-N.V.-Q.)
- Correspondence: (I.D.-G.); (A.F.); Tel.: +33-1-5727-7965 (I.D.-G.); +33-1-5727-7959 (A.F.)
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, 75013 Paris, France; (J.M.); (R.-N.V.-Q.)
- Reference Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, APHP, Institute of Myology, 75013 Paris, France
- Correspondence: (I.D.-G.); (A.F.); Tel.: +33-1-5727-7965 (I.D.-G.); +33-1-5727-7959 (A.F.)
| |
Collapse
|
30
|
Rosano KK, Wegner DJ, Shinawi M, Baldridge D, Bucelli RC, Dahiya S, White FV, Willing MC, McAllister W, Taft RJ, Bluske K, Buchanan A, Cole FS, Wambach JA. Biallelic ASCC1 variants including a novel intronic variant result in expanded phenotypic spectrum of spinal muscular atrophy with congenital bone fractures 2 (SMABF2). Am J Med Genet A 2021; 185:2190-2197. [PMID: 33931933 DOI: 10.1002/ajmg.a.62219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 11/08/2022]
Abstract
Spinal muscular atrophy with congenital bone fractures 2 (SMABF2), a type of arthrogryposis multiplex congenita (AMC), is characterized by congenital joint contractures, prenatal fractures of long bones, and respiratory distress and results from biallelic variants in ASCC1. Here, we describe an infant with severe, diffuse hypotonia, congenital contractures, and pulmonary hypoplasia characteristic of SMABF2, with the unique features of cleft palate, small spleen, transverse liver, and pulmonary thromboemboli with chondroid appearance. This infant also had impaired coagulation with diffuse petechiae and ecchymoses which has only been reported in one other infant with AMC. Using trio whole genome sequencing, our proband was identified to have biallelic variants in ASCC1. Using deep next generation sequencing of parental cDNA, we characterized alteration of splicing encoded by the novel, maternally inherited ASCC1 variant (c.297-8 T > G) which provides a mechanism for functional pathogenicity. The paternally inherited ASCC1 variant is a rare nonsense variant (c.466C > T; p.Arg156*) that has been previously identified in one other infant with AMC. This report extends the phenotypic characteristics of ASCC1-associated AMC (SMABF2) and describes a novel intronic variant that partially disrupts RNA splicing.
Collapse
Affiliation(s)
- Kristen K Rosano
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel J Wegner
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marwan Shinawi
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dustin Baldridge
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert C Bucelli
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Frances V White
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marcia C Willing
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - William McAllister
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | - Francis Sessions Cole
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jennifer A Wambach
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
31
|
Nair D, Li D, Erdogan H, Yoon A, Harr MH, Bergant G, Peterlin B, Škrjanec Pušenjak M, Jayakar P, Pfundt R, Jansen S, McWalter K, Sidhu A, Saliganan S, Agolini E, Jacob A, Pasquier J, Arash R, Kahrizi K, Najmabadi H, Ropers HH, Bhoj EJ. Discovery of a neuromuscular syndrome caused by biallelic variants in ASCC3. HGG ADVANCES 2021; 2:100024. [PMID: 35047834 PMCID: PMC8756546 DOI: 10.1016/j.xhgg.2021.100024] [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: 09/14/2020] [Accepted: 01/13/2021] [Indexed: 11/11/2022] Open
Abstract
Activating Signal Cointegrator 1 Complex, Subunit 3 (ASCC3) is part of the four-part ASC-1 transcriptional cointegrator complex. This complex includes ASCC1 (associated with spinal muscular atrophy with congenital bone fractures 2), TRIP4 (associated with spinal muscular atrophy with congenital bone fractures 1), and ASCC2 (not yet associated with human disease.) ASCC3 encodes a DNA helicase responsible for generating single-stranded DNA as part of the DNA damage response. Interestingly, ASCC3 expresses coding and non-coding isoforms, which act in opposition to balance the recovery of gene transcription after UV-induced DNA damage. Here we report the discovery of ASCC3 as the cause of a neuromuscular syndrome in seven unreported individuals from six unrelated families and updates on the one previously reported family. All the individuals share a neurologic phenotype that ranges from severe developmental delay to muscle fatigue. There appears to be genotype-phenotype correlation, as the most mildly affected individual is homozygous for a rare missense variant, while the more severely affected individuals are compound heterozygotes for a missense and a presumed loss-of-function (LOF) variant. There are no individuals with biallelic presumed LOF variants in our cohort or in gnomAD, as this genotype may not be compatible with life. In summary we report a syndrome in these eleven individuals from seven families with biallelic variants in ASCC3.
Collapse
|
32
|
Cho HW, Jin HS, Eom YB. Association between non-Caucasian-specific ASCC1 gene polymorphism and osteoporosis and obesity in Korean postmenopausal women. J Bone Miner Metab 2020; 38:868-877. [PMID: 32653958 DOI: 10.1007/s00774-020-01120-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/13/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Osteoporosis is a common disorder characterized by decreased bone mineral density (BMD). Interestingly, osteoporosis and obesity have several similar features, including a genetic predisposition and a common bone marrow stem cell. With aging, the composition of bone marrow shifts to adipocytes, osteoclast activity increases, and osteoblast function declines, resulting in osteoporosis. MATERIALS AND METHODS We performed a genome-wide association study (GWAS) analysis with osteoporosis and body mass index (BMI) and did identify an association in 349 and 384 SNPs by filtering with the significant p values (p < 0.001) of BMI and osteoporosis, respectively. RESULTS Only three of those SNPs were common (rs2326365, rs7097028, and rs11000205) between the SNPs significantly associated with BMI and/or osteoporosis in Korean Association REsource (KARE) females. Two of the three SNPs belonged to the ASCC1 gene and one to the FAM50B gene. We carried out a minor allele frequency (MAF) analysis of the rs7097028 and rs11000205 SNPs in the ASCC1 gene with a geographic genome variant browser. Both rs7097028 and rs11000205 in the ASCC1 gene were seen mostly in African and Southeast Asian populations. CONCLUSIONS Our results suggest that the ASCC1 gene is a significant genetic factor for determining the risk for both osteoporosis and obesity in KARE postmenopausal females.
Collapse
Affiliation(s)
- Hye-Won Cho
- Department of Medical Sciences, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea
| | - Hyun-Seok Jin
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Chungnam, 31499, Republic of Korea
| | - Yong-Bin Eom
- Department of Medical Sciences, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam, 31538, Republic of Korea.
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, 22 Soonchunhyang-ro, Shinchang-myeon, Asan, Chungcheongnam, 31538, Republic of Korea.
| |
Collapse
|
33
|
Dahan-Oliel N, Collins J, Rauch D, Bukovy G, Hamdy R, Rauch F. Bone densities and bone geometry in children and adolescents with arthrogryposis. Bone 2020; 137:115454. [PMID: 32464275 DOI: 10.1016/j.bone.2020.115454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/07/2020] [Accepted: 05/23/2020] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To describe bone densitometry results using lumbar spine dual-energy X-ray absorptiometry and forearm peripheral quantitative computed tomography (pQCT) in children with arthrogryposis multiplex congenita (AMC). STUDY DESIGN Prospective study. RESULTS Lumbar spine areal bone mineral density (BMD) was measured in 58 participants (mean age 6.8 years, range 1 month to 19.7 years; 26 males). The diagnostic subgroup was Amyoplasia in 27 participants, distal arthrogryposis (unclassified, n = 13; type 2A, n = 1; type 2B, n = 2; type 8, n = 2) in 18 patients, an unclassified form of arthrogryposis in 6 patients, and a syndromic form of arthrogryposis in 7 patients. The mean lumbar spine areal BMD was -0.4 (SD: 1.5) which was significantly below 0 (p < 0.05, one-sample t-test). The mean lumbar spine bone mineral apparent density z-score (+0.4 [SD: 1.4]), a measure that is largely independent of bone size, was not significantly different from 0 (P > 0.05). A subset of 22 patients aged 6 years or older (mean age 10.9 years, 11 males) had forearm pQCT analysis. Mean z-scores for trabecular and cortical volumetric BMD at the radius were similar to healthy controls. Radius periosteal bone circumference and bone mineral content were appropriate for height. These densitometric results did not differ between patients with Amyoplasia or individuals with other diagnoses. CONCLUSIONS Low areal BMD in children and adolescents with AMC reflects their smaller bone size rather than a specific bone mass deficit. These data do not suggest that children and adolescents with AMC in general require regular monitoring by bone densitometry unless there are specific clinical concerns.
Collapse
Affiliation(s)
- Noemi Dahan-Oliel
- Shriners Hospital for Children, Montreal, Quebec, Canada; School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | - Damian Rauch
- Shriners Hospital for Children, Montreal, Quebec, Canada
| | - Gloria Bukovy
- Shriners Hospital for Children, Montreal, Quebec, Canada
| | - Reggie Hamdy
- Shriners Hospital for Children, Montreal, Quebec, Canada; Faculty of Medicine, Division of Orthopaedic Surgery, McGill University, Montreal, Quebec, Canada
| | - Frank Rauch
- Shriners Hospital for Children, Montreal, Quebec, Canada; Department of Pediatrics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
34
|
Tsai CL, Tsai CN, Lee YS, Wang HS, Lee LY, Lin CY, Yang SY, Chao A. Genetic analysis of a Taiwanese family identifies a DMRT3-OAS3 interaction that is involved in human sexual differentiation through the regulation of ESR1 expression. Fertil Steril 2020; 114:133-143. [PMID: 32553473 DOI: 10.1016/j.fertnstert.2020.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To identify the genetic etiology of recurrent disorders of sex development (DSDs) in a Taiwanese family with 46,XY sex reversal and hypospadias. DESIGN Genetic and functional studies. SETTING Academic hospital. PATIENT(S) A three-generation family consisting of 22 members, with eight cases of 46,XY DSD, of whom four have 46,XY male-to-female sex reversal and four are 46,XY males with hypospadias. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Results of exome sequencing and in vitro protein and RNA analyses. RESULT(S) All patients with DSDs were found to carry heterozygous missense mutations in the doublesex and mab-3-related transcription factor 3 (DMRT3; rs187176004, c.A815C, p.K272T) and 2',5'-oligoadenylate synthetase 3 (OAS3; rs16942374, c.G2606A, p.R869H) genes. The DMRT3 mutation increased estrogen receptor 1 (ESR1) expression. Upon binding with the OAS3-RNase L complex, wild-type DMRT3 promoted degradation of ESR1 mRNA. However, the DMRT3A815C-OAS3G2606A complex interacted less strongly with ESR1 mRNA and RNase L, ultimately preventing ESR1 mRNA degradation. The interactions between DMRT3, OAS3, and RNase L were confirmed in the patients' testis. CONCLUSION(S) Our results indicate that DMRT3 and OAS3 are involved in human DSDs by controlling ESR1 expression.
Collapse
Affiliation(s)
- Chia-Lung Tsai
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chi-Neu Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yun-Shien Lee
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan
| | - Hsin-Shih Wang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chiao-Yun Lin
- Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu Yuan Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
35
|
Zhou Z, Li H, Li H, Zhang J, Fu K, Cao C, Deng F, Luo J. Comprehensive analysis of the differential expression profile of microRNAs in rats with spinal cord injury treated by electroacupuncture. Mol Med Rep 2020; 22:751-762. [PMID: 32468009 PMCID: PMC7339738 DOI: 10.3892/mmr.2020.11161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 04/22/2020] [Indexed: 01/05/2023] Open
Abstract
Abnormal microRNA (miRNA) expression has been implicated in spinal cord injury (SCI), but the underlying mechanisms are poorly understood. To observe the effect of electroacupuncture (EA) on miRNA expression profiles in SCI rats and investigate the potential mechanisms involved in this process, Sprague-Dawley rats were divided into sham, SCI and SCI+EA groups (n=6 each). Basso, Beattie and Bresnahan (BBB) scoring and hematoxylin-eosin staining of cortical tissues were used to evaluate spinal cord recovery with EA treatment 21 days post-surgery across the three groups. To investigate miRNA expression profiles, 6 Sprague-Dawley rats were randomly divided into SCI and SCI+EA groups (n=3 in each group) and examined using next-generation sequencing. Integrated miRNA-mRNA-pathway network analysis was performed to elucidate the interaction network of the candidate miRNAs, their target genes and the involved pathways. Behavioral scores suggested that hindlimb motor functions improved with EA treatments. Apoptotic indices were lower in the SCI+EA group compared with the SCI group. It was also observed that 168 miRNAs were differentially expressed between the SCI and SCI+EA groups, with 29 upregulated and 139 downregulated miRNAs in the SCI+EA group. Changes in miRNA expression are involved in SCI physiopathology, including inflammation and apoptosis. Reverse transcription-quantitative PCR measurement of the five candidate miRNAs, namely rno-miR-219a-5p, rno-miR-486, rno-miR-136-5p, rno-miR-128-3p, and rno-miR-7b, was consistent with RNA sequencing data. Integrated miRNA-mRNA-pathway analysis suggested that the MAPK, Wnt and NF-κB signaling pathways were involved in EA-mediated recovery from SCI. The present study evaluated the miRNA expression profiles involved in EA-treated SCI rats and demonstrated the potential mechanism and functional role of miRNAs in SCI in rats.
Collapse
Affiliation(s)
- Zhidong Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Hejian Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Hongchun Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Kaiwen Fu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Cao Cao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Fumou Deng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Jun Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
36
|
Li G, Zhang H, Wu J, Wang A, Yang F, Chen B, Gao Y, Ma X, Xu Y. Hepcidin deficiency causes bone loss through interfering with the canonical Wnt/β-catenin pathway via Forkhead box O3a. J Orthop Translat 2020; 23:67-76. [PMID: 32514392 PMCID: PMC7267010 DOI: 10.1016/j.jot.2020.03.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/01/2020] [Accepted: 03/23/2020] [Indexed: 01/05/2023] Open
Abstract
Objective Hepcidin deficiency is known to cause body iron accumulation and bone microarchitecture defects, but the exact underlying mechanisms of hepcidin deficiency-induced bone loss remain unclear. Our objective was to understand the molecular mechanism of hepcidin deficiency-induced bone loss. Methods The bone phenotypes of wild type (WT) and hepcidin knockout (Hepcidin-KO) mice were measured by microcomputed tomography. The osteoclastic marker of the bone was measured by tartrate-resistant acid phosphatase staining. The osteoblastic marker of the bone was measured by immunostaining of osteocalcin. Primary osteoblastic and osteoclastic differentiation was performed using bone marrow cells. The mature osteoclast was determined by tartrate-resistant acid phosphatase staining, pit formation assay and relative gene expression. The mature osteoblast was determined by alkaline phosphatase activity, alkaline phosphatase staining, Alizarin Red staining and relative gene expression. The protein expression of β-catenin, TCF4/TCF7L2 and Forkhead box O3a (FOXO3a) was measured by Western blot and their combination by co-immunoprecipitation. In vivo study was performed by tail vein administration of FOXO3a-RNAi using an adeno-associated virus in Hepcidin-KO mice. Results We found that Hepcidin-KO mice exhibited iron accumulation and bone loss compared with WT mice. The osteoclastic differentiation of bone marrow-derived macrophages from Hepcidin-KO mice was not significantly different from that of bone marrow–derived macrophages from WT mice. However, the osteoblastic differentiation of bone marrow–derived mesenchymal stem cells from Hepcidin-KO mice was obviously decreased compared with that of bone marrow–derived mesenchymal stem cells from WT mice. Furthermore, it was confirmed in this study that upon hepcidin deficiency, β-catenin, TCF4/TCF7L2 and FOXO3a expression in bone tissues was not altered, but β-catenin combination with TCF4/TCF7L2 was strongly inhibited by β-catenin combination with FOXO3a, indicating that the canonical Wnt/β-catenin pathway was affected. Tail vein administration of FOXO3a-RNAi using an adeno-associated virus in Hepcidin-KO mice resulted in bone mass recovery. Conclusion These findings suggested that hepcidin deficiency might cause bone loss by interfering with the canonical Wnt/β-catenin pathway via FOXO3a, and FOXO3a inhibition would be a possible approach to treat hepcidin deficiency-induced bone loss. The translational potential of this article Hepcidin deficiency, as well as iron accumulation, has been considered as a risk factor for osteoporosis. For this kind of osteoporosis, inhibition of FOXO3a either by neutralized antibody or AAV-mediated RNAi, represents an effective and promising method.
Collapse
Affiliation(s)
- Guangfei Li
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Hui Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Jiadong Wu
- Department of Orthopaedics, The Affiliated Yancheng Hospital of Southeast University Medical College, 224005, Yancheng, China
| | - Aifei Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Fan Yang
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Bin Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Yan Gao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| | - Xiaowei Ma
- Department of Orthopaedics, Zhongshan Hospital of Dalian University, 116001, Dalian, China
| | - Youjia Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.,Osteoporosis Institute of Soochow University, 1055 Sanxiang Road, 215004, Suzhou, China
| |
Collapse
|
37
|
Lu W, Liang M, Su J, Wang J, Li L, Zhang S, Qin Z, Huang L, Lu Y, Yi S, Yi S, Xie B, Zheng H, Luo J, Gao X, Shen Y. Novel compound heterozygous pathogenic variants in ASCC1 in a Chinese patient with spinal muscular atrophy with congenital bone fractures 2 : Evidence supporting a "Definitive" gene-disease relationship. Mol Genet Genomic Med 2020; 8:e1212. [PMID: 32160656 PMCID: PMC7216800 DOI: 10.1002/mgg3.1212] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/01/2020] [Accepted: 02/24/2020] [Indexed: 12/21/2022] Open
Abstract
Background A very limited spectrum of ASCC1 pathogenic variants had been reported in six (mostly consanguineous) families with spinal muscular atrophy with congenital bone fractures 2 [OMIM #616867] since 2016. Methods A proband from a non‐consanguineous Chinese family presented with neonatal severe hypotonia, respiratory distress, muscle weakness, and atrophy, as well as congenital bone fractures was performed by exome sequencing. Results A compound heterozygosity of a nonsense (c.932C>G,p.Ser311Ter) and an exon 5 deletion in ASCC1 segregating with phenotypes was detected, both variants are novel and pathogenic. Since ASCC1 is a relatively new disease gene, we performed the gene curation by ClinGen SOP. The existing evidence is sufficient to support a "Definitive" level of disease‐gene relationship. Conclusion This case report expended the mutation spectrum of ASCC1 and support the notion that this novel disease also occurs in outbreed populations and this is a rare disease but may still be underdiagnosed due to its perinatal lethal outcomes.
Collapse
Affiliation(s)
- Weiliang Lu
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Mingxing Liang
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jiasun Su
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jin Wang
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lingxiao Li
- Department of Neonatology, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shujie Zhang
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zailong Qin
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Limei Huang
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yingchi Lu
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shang Yi
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Sheng Yi
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - BoBo Xie
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Haiyang Zheng
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jingsi Luo
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaoyan Gao
- Department of Neonatology, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yiping Shen
- Genetic and Metabolic Central Laboratory, Birth Defect Prevention Research Institute, Maternal and Child Health Hospital, Children's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.,Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Vallerga CL, Zhang F, Fowdar J, McRae AF, Qi T, Nabais MF, Zhang Q, Kassam I, Henders AK, Wallace L, Montgomery G, Chuang YH, Horvath S, Ritz B, Halliday G, Hickie I, Kwok JB, Pearson J, Pitcher T, Kennedy M, Bentley SR, Silburn PA, Yang J, Wray NR, Lewis SJG, Anderson T, Dalrymple-Alford J, Mellick GD, Visscher PM, Gratten J. Analysis of DNA methylation associates the cystine-glutamate antiporter SLC7A11 with risk of Parkinson's disease. Nat Commun 2020; 11:1238. [PMID: 32144264 PMCID: PMC7060318 DOI: 10.1038/s41467-020-15065-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 02/17/2020] [Indexed: 11/09/2022] Open
Abstract
An improved understanding of etiological mechanisms in Parkinson's disease (PD) is urgently needed because the number of affected individuals is projected to increase rapidly as populations age. We present results from a blood-based methylome-wide association study of PD involving meta-analysis of 229 K CpG probes in 1,132 cases and 999 controls from two independent cohorts. We identify two previously unreported epigenome-wide significant associations with PD, including cg06690548 on chromosome 4. We demonstrate that cg06690548 hypermethylation in PD is associated with down-regulation of the SLC7A11 gene and show this is consistent with an environmental exposure, as opposed to medications or genetic factors with effects on DNA methylation or gene expression. These findings are notable because SLC7A11 codes for a cysteine-glutamate anti-porter regulating levels of the antioxidant glutathione, and it is a known target of the environmental neurotoxin β-methylamino-L-alanine (BMAA). Our study identifies the SLC7A11 gene as a plausible biological target in PD.
Collapse
Affiliation(s)
- Costanza L Vallerga
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Futao Zhang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Javed Fowdar
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Brisbane, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ting Qi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Marta F Nabais
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,University of Exeter Medical School, Exeter EX2 5DW, Devon, UK
| | - Qian Zhang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Irfahan Kassam
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Grant Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Yu-Hsuan Chuang
- Department of Epidemiology, Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA.,Department of Biostatistics, Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, UCLA, Los Angeles, CA, USA.,Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.,Department of Environmental Health, Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Glenda Halliday
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Ian Hickie
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - John B Kwok
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - John Pearson
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Toni Pitcher
- New Zealand Brain Research Institute, Christchurch, New Zealand.,Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Martin Kennedy
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Steven R Bentley
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Brisbane, Australia
| | - Peter A Silburn
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Simon J G Lewis
- Brain and Mind Centre & Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Tim Anderson
- New Zealand Brain Research Institute, Christchurch, New Zealand.,Department of Medicine, University of Otago, Christchurch, New Zealand
| | - John Dalrymple-Alford
- New Zealand Brain Research Institute, Christchurch, New Zealand.,Department of Psychology, University of Canterbury, Christchurch, New Zealand
| | - George D Mellick
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Brisbane, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia. .,Queensland Brain Institute, The University of Queensland, Brisbane, Australia.
| | - Jacob Gratten
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia. .,Mater Research Institute, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
39
|
Vester K, Santos KF, Kuropka B, Weise C, Wahl MC. The inactive C-terminal cassette of the dual-cassette RNA helicase BRR2 both stimulates and inhibits the activity of the N-terminal helicase unit. J Biol Chem 2019; 295:2097-2112. [PMID: 31914407 DOI: 10.1074/jbc.ra119.010964] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/27/2019] [Indexed: 11/06/2022] Open
Abstract
The RNA helicase bad response to refrigeration 2 homolog (BRR2) is required for the activation of the spliceosome before the first catalytic step of RNA splicing. BRR2 represents a distinct subgroup of Ski2-like nucleic acid helicases whose members comprise tandem helicase cassettes. Only the N-terminal cassette of BRR2 is an active ATPase and can unwind substrate RNAs. The C-terminal cassette represents a pseudoenzyme that can stimulate RNA-related activities of the N-terminal cassette. However, the molecular mechanisms by which the C-terminal cassette modulates the activities of the N-terminal unit remain elusive. Here, we show that N- and C-terminal cassettes adopt vastly different relative orientations in a crystal structure of BRR2 in complex with an activating domain of the spliceosomal Prp8 protein at 2.4 Å resolution compared with the crystal structure of BRR2 alone. Likewise, inspection of BRR2 structures within spliceosomal complexes revealed that the cassettes occupy different relative positions and engage in different intercassette contacts during different splicing stages. Engineered disulfide bridges that locked the cassettes in two different relative orientations had opposite effects on the RNA-unwinding activity of the N-terminal cassette, with one configuration enhancing and the other configuration inhibiting RNA unwinding compared with the unconstrained protein. Moreover, we found that differences in relative positioning of the cassettes strongly influence RNA-stimulated ATP hydrolysis by the N-terminal cassette. Our results indicate that the inactive C-terminal cassette of BRR2 can both positively and negatively affect the activity of the N-terminal helicase unit from a distance.
Collapse
Affiliation(s)
- Karen Vester
- Structural Biochemistry Group, Department of Biochemistry, Freie Universität Berlin, Takustrasse 63, D-14195 Berlin, Germany
| | - Karine F Santos
- Structural Biochemistry Group, Department of Biochemistry, Freie Universität Berlin, Takustrasse 63, D-14195 Berlin, Germany
| | - Benno Kuropka
- Protein Biochemistry Group, Department of Biochemistry, Freie Universität Berlin, Thielallee 63, D-14195 Berlin, Germany
| | - Christoph Weise
- Protein Biochemistry Group, Department of Biochemistry, Freie Universität Berlin, Thielallee 63, D-14195 Berlin, Germany
| | - Markus C Wahl
- Structural Biochemistry Group, Department of Biochemistry, Freie Universität Berlin, Takustrasse 63, D-14195 Berlin, Germany; Macromolecular Crystallography Group, Helmholtz-Zentrum Berlin für Materialien und Energie, Albert-Einstein-Strasse 15, D-12489 Berlin, Germany.
| |
Collapse
|
40
|
Villar-Quiles RN, Catervi F, Cabet E, Juntas-Morales R, Genetti CA, Gidaro T, Koparir A, Yüksel A, Coppens S, Deconinck N, Pierce-Hoffman E, Lornage X, Durigneux J, Laporte J, Rendu J, Romero NB, Beggs AH, Servais L, Cossée M, Olivé M, Böhm J, Duband-Goulet I, Ferreiro A. ASC-1 Is a Cell Cycle Regulator Associated with Severe and Mild Forms of Myopathy. Ann Neurol 2019; 87:217-232. [PMID: 31794073 DOI: 10.1002/ana.25660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Recently, the ASC-1 complex has been identified as a mechanistic link between amyotrophic lateral sclerosis and spinal muscular atrophy (SMA), and 3 mutations of the ASC-1 gene TRIP4 have been associated with SMA or congenital myopathy. Our goal was to define ASC-1 neuromuscular function and the phenotypical spectrum associated with TRIP4 mutations. METHODS Clinical, molecular, histological, and magnetic resonance imaging studies were made in 5 families with 7 novel TRIP4 mutations. Fluorescence activated cell sorting and Western blot were performed in patient-derived fibroblasts and muscles and in Trip4 knocked-down C2C12 cells. RESULTS All mutations caused ASC-1 protein depletion. The clinical phenotype was purely myopathic, ranging from lethal neonatal to mild ambulatory adult patients. It included early onset axial and proximal weakness, scoliosis, rigid spine, dysmorphic facies, cutaneous involvement, respiratory failure, and in the older cases, dilated cardiomyopathy. Muscle biopsies showed multiminicores, nemaline rods, cytoplasmic bodies, caps, central nuclei, rimmed fibers, and/or mild endomysial fibrosis. ASC-1 depletion in C2C12 and in patient-derived fibroblasts and muscles caused accelerated proliferation, altered expression of cell cycle proteins, and/or shortening of the G0/G1 cell cycle phase leading to cell size reduction. INTERPRETATION Our results expand the phenotypical and molecular spectrum of TRIP4-associated disease to include mild adult forms with or without cardiomyopathy, associate ASC-1 depletion with isolated primary muscle involvement, and establish TRIP4 as a causative gene for several congenital muscle diseases, including nemaline, core, centronuclear, and cytoplasmic-body myopathies. They also identify ASC-1 as a novel cell cycle regulator with a key role in cell proliferation, and underline transcriptional coregulation defects as a novel pathophysiological mechanism. ANN NEUROL 2020;87:217-232.
Collapse
Affiliation(s)
- Rocío N Villar-Quiles
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, Paris, France.,Reference Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, APHP, Institute of Myology, Paris, France
| | - Fabio Catervi
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, Paris, France
| | - Eva Cabet
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, Paris, France
| | - Raul Juntas-Morales
- Neuromuscular Unit, University Hospital Center Montpellier/EA7402 University of Montpellier, University Institute of Clinical Research, Montpellier, France
| | - Casie A Genetti
- Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | | | - Asuman Koparir
- Department of Molecular Biology and Genetics, Biruni University, Istanbul, Turkey
| | - Adnan Yüksel
- Department of Molecular Biology and Genetics, Biruni University, Istanbul, Turkey
| | - Sandra Coppens
- Department of Pediatric Neurology, Reference Neuromuscular Center, Queen Fabiola Children's University Hospital, Free University of Brussels, Brussels, Belgium
| | - Nicolas Deconinck
- Department of Pediatric Neurology, Reference Neuromuscular Center, Queen Fabiola Children's University Hospital, Free University of Brussels, Brussels, Belgium
| | - Emma Pierce-Hoffman
- Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Xavière Lornage
- Department of Translational Medicine and Neurogenetics, Institute of Genetics and Molecular and Cellular Biology, National Institute of Health and Medical Research U1258, National Center for Scientific Research UMR7104, University of Strasbourg, Illkirch, France
| | - Julien Durigneux
- Department of Neuropediatrics, University Hospital Center Angers, Neuromuscular Diseases Reference Center Antlantique Occitanie Caraïbe, Angers, France
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institute of Genetics and Molecular and Cellular Biology, National Institute of Health and Medical Research U1258, National Center for Scientific Research UMR7104, University of Strasbourg, Illkirch, France
| | - John Rendu
- Laboratory of Biochemistry and Molecular Genetics, University Hospital Center Grenoble, Grenoble, France
| | - Norma B Romero
- Reference Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, APHP, Institute of Myology, Paris, France.,Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Paris, France
| | - Alan H Beggs
- Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Laurent Servais
- I-Motion, Institute of Myology, APHP, Paris, France.,Division of Child Neurology, Neuromuscular Diseases Reference Center, Department of Pediatrics, Liège University Hospital and University of Liège, Liège, Belgium
| | - Mireille Cossée
- Molecular Genetics Laboratory, University Hospital Center Montpellier/National Institute of Health and Medical Research U827, University Institute of Clinical Research, Montpellier, France
| | - Montse Olivé
- Neuropathology Unit, Department of Pathology and Neuromuscular Unit, Institute of Biomedical Research of Bellvitge-University Hospital of Bellvitge, Barcelona, Spain
| | - Johann Böhm
- Department of Translational Medicine and Neurogenetics, Institute of Genetics and Molecular and Cellular Biology, National Institute of Health and Medical Research U1258, National Center for Scientific Research UMR7104, University of Strasbourg, Illkirch, France
| | - Isabelle Duband-Goulet
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, Paris, France
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, UMR8251, University of Paris/National Center for Scientific Research, Paris, France.,Reference Center for Neuromuscular Disorders, Pitié-Salpêtrière Hospital, APHP, Institute of Myology, Paris, France
| |
Collapse
|
41
|
Giuffrida MG, Mastromoro G, Guida V, Truglio M, Fabbretti M, Torres B, Mazza T, De Luca A, Roggini M, Bernardini L, Pizzuti A. A new case of SMABF2 diagnosed in stillbirth expands the prenatal presentation and mutational spectrum of ASCC1. Am J Med Genet A 2019; 182:508-512. [PMID: 31880396 DOI: 10.1002/ajmg.a.61431] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 12/24/2022]
Abstract
Spinal muscular atrophy with congenital bone fractures 2 (SMABF2) is a rare autosomal recessive neuromuscular disorder characterized by arthrogryposis multiplex congenita and prenatal fractures of the long bones, with poor prognosis. The most affected patients present with biallelic loss-of-function nucleotide variants in ASCC1 gene, coding a subunit of the transcriptional coactivator ASC-1 complex, although the exact pathogenesis is yet unknown. This work describes the first case of SMABF2 in a stillbirth with documented evolution of the disease in the prenatal period. A microdeletion copy number variant (CNV) of about 64 Kb, involving four exons of ASCC1, was firstly detected by microarray analysis, requested for arthrogryposis and hydrops. Subsequent exome analysis disclosed a nucleotide variant of the same gene [c.1027C>T; (p. Arg343*)], resulting in the introduction of a premature termination codon. This stillbirth represents the first case of ASCC1 compound heterozygosity, due to an exonic microdeletion and a nucleotide variant, expanding the mutational spectrum of this gene. It also provides further evidence that exonic CNVs are an underestimated cause of disease-alleles and that the integrated use of the last generation genetic analysis tools, together with careful clinical evaluations, are fundamental for the characterization of rare diseases even in the prenatal setting.
Collapse
Affiliation(s)
- Maria G Giuffrida
- Medical Genetics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Gioia Mastromoro
- Department of Experimental Medicine, Sapienza University, Policlinico Umberto I Hospital, Rome, Italy
| | - Valentina Guida
- Medical Genetics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Mauro Truglio
- Bioinformatics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Maria Fabbretti
- Medical Genetics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Barbara Torres
- Medical Genetics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Alessandro De Luca
- Medical Genetics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Mario Roggini
- Pediatrics and Child Neuropsychiatry Department, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Laura Bernardini
- Medical Genetics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy
| | - Antonio Pizzuti
- Medical Genetics Unit, Casa Sollievo della Sofferenza IRCCS Foundation, San Giovanni Rotondo, Italy.,Department of Experimental Medicine, Sapienza University, Policlinico Umberto I Hospital, Rome, Italy
| |
Collapse
|
42
|
Vorstman JAS, Olde Loohuis LM, Kahn RS, Ophoff RA. Double hits in schizophrenia. Hum Mol Genet 2019; 27:2755-2761. [PMID: 29767709 DOI: 10.1093/hmg/ddy175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/30/2018] [Accepted: 05/04/2018] [Indexed: 11/14/2022] Open
Abstract
The co-occurrence of a copy number variant (CNV) and a functional variant on the other allele may be a relevant genetic mechanism in schizophrenia. We hypothesized that the cumulative burden of such double hits-in particular those composed of a deletion and a coding single-nucleotide variation (SNV)-is increased in patients with schizophrenia. We combined CNV data with coding variants data in 795 patients with schizophrenia and 474 controls. To limit false CNV-detection, only CNVs called by two algorithms were included. CNV-affected genes were subsequently examined for coding SNVs, which we termed "CNV-SNVs." Correcting for total queried sequence, we assessed the CNV-SNV-burden and the combined predicted deleterious effect. We estimated P-values by permutation of the phenotype. We detected 105 CNV-SNVs; 67 in duplicated and 38 in deleted genic sequence. Although the difference in CNV-SNVs rates was not significant, the combined deleteriousness inferred by CNV-SNVs in deleted sequence was almost 4-fold higher in cases compared with controls (nominal P = 0.009). This effect may be driven by a higher number of CNV-SNVs and/or by a higher degree of predicted deleteriousness of CNV-SNVs. No such effect was observed for duplications. We provide early evidence that deletions co-occurring with a functional variant may be relevant, albeit of modest impact, for the genetic etiology of schizophrenia. Large-scale consortium studies are required to validate our findings. Sequence-based analyses would provide the best resolution for detection of CNVs as well as coding variants genome-wide.
Collapse
Affiliation(s)
- Jacob A S Vorstman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Psychiatry, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada.,Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, CA, USA
| | | | - René S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Psychiatry, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Roel A Ophoff
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, CA, USA.,Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
43
|
Iolascon G, Paoletta M, Liguori S, Curci C, Moretti A. Neuromuscular Diseases and Bone. Front Endocrinol (Lausanne) 2019; 10:794. [PMID: 31824418 PMCID: PMC6886381 DOI: 10.3389/fendo.2019.00794] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/30/2019] [Indexed: 02/05/2023] Open
Abstract
Neuromuscular diseases (NMDs) are inherited or acquired conditions affecting skeletal muscles, motor nerves, or neuromuscular junctions. Most of them are characterized by a progressive damage of muscle fibers with reduced muscle strength, disability, and poor health-related quality of life of affected patients. In this scenario, skeletal health is usually compromised as a consequence of modified bone-muscle cross-talk including biomechanical and bio-humoral issues, resulting in increased risk of bone fragility and fractures. In addition, NMD patients frequently face nutritional issues, including malnutrition due to feeding disorders and swallowing problems that might affect bone health. Moreover, in these patients, low levels of physical activity or immobility are common and might lead to overweight or obesity that can also interfere with bone strength features. Also, vitamin D deficiency could play a critical role both in the pathogenesis and in the clinical scenario of many NMDs, suggesting that its correction could be useful in maintaining or enhancing bone health, especially in the early phases of NMDs. Last but not least, specific disease-modifying drugs, available for some NMDs, are frequently burdened with adverse effects on bone tissue. For example, glucocorticoid therapy, standard of care for many muscular dystrophies, prolongs long-term survival in treated patients; nevertheless, high dose and/or chronic use of these drugs are a common cause of secondary osteoporosis. This review addresses the current state of knowledge about the factors that play a role in determining bone alterations reported in NMDs, how these factors can modify the biological pathways underlying bone health, and which are the available interventions to manage bone involvement in patients affected by NMDs. Considering the complexity of care of these patients, an interdisciplinary and multimodal management strategy based on both pharmacological and non-pharmacological interventions is recommended, particularly targeting musculoskeletal issues that are closely related to functional independence as well as social implications.
Collapse
Affiliation(s)
- Giovanni Iolascon
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | | | | | | | | |
Collapse
|
44
|
Mallam AL, Sae-Lee W, Schaub JM, Tu F, Battenhouse A, Jang YJ, Kim J, Wallingford JB, Finkelstein IJ, Marcotte EM, Drew K. Systematic Discovery of Endogenous Human Ribonucleoprotein Complexes. Cell Rep 2019; 29:1351-1368.e5. [PMID: 31665645 PMCID: PMC6873818 DOI: 10.1016/j.celrep.2019.09.060] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/30/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022] Open
Abstract
RNA-binding proteins (RBPs) play essential roles in biology and are frequently associated with human disease. Although recent studies have systematically identified individual RNA-binding proteins, their higher-order assembly into ribonucleoprotein (RNP) complexes has not been systematically investigated. Here, we describe a proteomics method for systematic identification of RNP complexes in human cells. We identify 1,428 protein complexes that associate with RNA, indicating that more than 20% of known human protein complexes contain RNA. To explore the role of RNA in the assembly of each complex, we identify complexes that dissociate, change composition, or form stable protein-only complexes in the absence of RNA. We use our method to systematically identify cell-type-specific RNA-associated proteins in mouse embryonic stem cells and finally, distribute our resource, rna.MAP, in an easy-to-use online interface (rna.proteincomplexes.org). Our system thus provides a methodology for explorations across human tissues, disease states, and throughout all domains of life.
Collapse
Affiliation(s)
- Anna L Mallam
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Wisath Sae-Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Jeffrey M Schaub
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Fan Tu
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Anna Battenhouse
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Yu Jin Jang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Ilya J Finkelstein
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Edward M Marcotte
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Kevin Drew
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
45
|
Dieterich K, Kimber E, Hall JG. Central nervous system involvement in arthrogryposis multiplex congenita: Overview of causes, diagnosis, and care. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2019; 181:345-353. [PMID: 31410997 DOI: 10.1002/ajmg.c.31732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/13/2019] [Accepted: 07/17/2019] [Indexed: 12/14/2022]
Abstract
Arthrogryposis or AMC, arthrogryposis multiplex congenita, is defined as multiple congenital joint contractures in more than two joints and in different body areas. The common cause of all AMC is lack of movement in utero, which in turn can have different causes, one of which is CNS involvement. Intellectual disability/CNS involvement is found in approximately 25% of all AMC. AMC with CNS involvement includes a large number of genetic syndromes. So far, more than 400 genes have been identified as linked to AMC, with and without CNS involvement. A number of neonatally lethal syndromes and syndromes resulting in severe disability due to CNS malfunction belong to this group of syndromes. There are several X-linked disorders with AMC, which are primarily related to intellectual disability. A number of neuromuscular disorders may include AMC and CNS/brain involvement. Careful clinical evaluation by a geneticist and a pediatrician/pediatric neurologist is the first step in making a specific diagnosis. Further investigations may include MRI of the brain and spinal cord, electroencephalogram, blood chemistry for muscle enzymes, other organ investigations (ophtalmology, cardiology, gastrointestinal, and genitourinary systems). Nerve conduction studies, electromyogram, and muscle pathology may be of help when there is associated peripheral nervous system involvement. But most importantly, genetic investigations with targeted or rather whole exome or genome sequencing should be performed. A correct diagnosis is important in planning adequate treatment, in genetic counselling and also for future understanding of pathogenic mechanisms and possible new treatments. A multidiciplinary team is needed both in investigation and treatment.
Collapse
Affiliation(s)
- Klaus Dieterich
- Univ. Grenoble Alpes, Inserm, U1216, GIN, Grenoble, France.,CHU Grenoble Alpes, Génétique Médicale, Grenoble, France
| | - Eva Kimber
- Department of Women's and Children's Health, Uppsala University Children's Hospital, Uppsala, Sweden.,Department of Paediatrics, Institute of Clinical Sciences, University of Gothenburg, The Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Judith G Hall
- Professor Emerita, Department of Pediatrics and Medical Genetics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
46
|
Chi B, O'Connell JD, Iocolano AD, Coady JA, Yu Y, Gangopadhyay J, Gygi SP, Reed R. The neurodegenerative diseases ALS and SMA are linked at the molecular level via the ASC-1 complex. Nucleic Acids Res 2019; 46:11939-11951. [PMID: 30398641 PMCID: PMC6294556 DOI: 10.1093/nar/gky1093] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022] Open
Abstract
Understanding the molecular pathways disrupted in motor neuron diseases is urgently needed. Here, we employed CRISPR knockout (KO) to investigate the functions of four ALS-causative RNA/DNA binding proteins (FUS, EWSR1, TAF15 and MATR3) within the RNAP II/U1 snRNP machinery. We found that each of these structurally related proteins has distinct roles with FUS KO resulting in loss of U1 snRNP and the SMN complex, EWSR1 KO causing dissociation of the tRNA ligase complex, and TAF15 KO resulting in loss of transcription factors P-TEFb and TFIIF. However, all four ALS-causative proteins are required for association of the ASC-1 transcriptional co-activator complex with the RNAP II/U1 snRNP machinery. Remarkably, mutations in the ASC-1 complex are known to cause a severe form of Spinal Muscular Atrophy (SMA), and we show that an SMA-causative mutation in an ASC-1 component or an ALS-causative mutation in FUS disrupts association between the ASC-1 complex and the RNAP II/U1 snRNP machinery. We conclude that ALS and SMA are more intimately tied to one another than previously thought, being linked via the ASC-1 complex.
Collapse
Affiliation(s)
- Binkai Chi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| | - Jeremy D O'Connell
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| | - Alexander D Iocolano
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| | - Jordan A Coady
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| | - Yong Yu
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| | - Jaya Gangopadhyay
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| | - Robin Reed
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston MA 02115, USA
| |
Collapse
|
47
|
Böhm J, Malfatti E, Oates E, Jones K, Brochier G, Boland A, Deleuze JF, Romero NB, Laporte J. Novel ASCC1 mutations causing prenatal-onset muscle weakness with arthrogryposis and congenital bone fractures. J Med Genet 2018; 56:617-621. [PMID: 30327447 DOI: 10.1136/jmedgenet-2018-105390] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/03/2018] [Accepted: 09/22/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND The activating signal cointegrator 1 (ASC-1) complex acts as a transcriptional coactivator for a variety of transcription factors and consists of four subunits: ASCC1, ASCC2, ASCC3 and TRIP4. A single homozygous mutation in ASCC1 has recently been reported in two families with a severe muscle and bone disorder. OBJECTIVE We aim to contribute to a better understanding of the ASCC1-related disorder. METHODS Here, we provide a clinical, histological and genetic description of three additional ASCC1 families. RESULTS All patients presented with severe prenatal-onset muscle weakness, neonatal hypotonia and arthrogryposis, and congenital bone fractures. The muscle biopsies from the affected infants revealed intense oxidative rims beneath the sarcolemma and scattered remnants of sarcomeres with enlarged Z-bands, potentially representing a histopathological hallmark of the disorder. Sequencing identified recessive nonsense or frameshift mutations in ASCC1, including two novel mutations. CONCLUSION Overall, this work expands the ASCC1 mutation spectrum, sheds light on the muscle histology of the disorder and emphasises the physiological importance of the ASC-1 complex in fetal muscle and bone development.
Collapse
Affiliation(s)
- Johann Böhm
- Departement of Translational Medicine and Neurogenetics, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Inserm U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Edoardo Malfatti
- Morphological Unit, Institut de Myologie, GHU La Pitié-Salpêtrière, Paris, France.,Centre de Référence de Pathologie Neuromusculaire Paris-Est, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Emily Oates
- Institute for Neuroscience and Muscle Research, Kid's Research Institute, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Kristi Jones
- Institute for Neuroscience and Muscle Research, Kid's Research Institute, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Guy Brochier
- Morphological Unit, Institut de Myologie, GHU La Pitié-Salpêtrière, Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de biologie François Jacob, CEA, Évry, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de biologie François Jacob, CEA, Évry, France
| | - Norma Beatriz Romero
- Morphological Unit, Institut de Myologie, GHU La Pitié-Salpêtrière, Paris, France.,Centre de Référence de Pathologie Neuromusculaire Paris-Est, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jocelyn Laporte
- Departement of Translational Medicine and Neurogenetics, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Inserm U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| |
Collapse
|
48
|
Beecroft SJ, Lombard M, Mowat D, McLean C, Cairns A, Davis M, Laing NG, Ravenscroft G. Genetics of neuromuscular fetal akinesia in the genomics era. J Med Genet 2018; 55:505-514. [PMID: 29959180 DOI: 10.1136/jmedgenet-2018-105266] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/22/2018] [Accepted: 04/19/2018] [Indexed: 12/27/2022]
Abstract
Fetal hypokinesia or akinesia encompasses a broad spectrum of disorders, united by impaired movement in utero. Often, the underlying aetiology is genetic in origin, affecting part of the neuromuscular system. The affordable and high-throughput nature of next-generation DNA sequencing has led to an explosion in disease gene discovery across rare diseases, including fetal akinesias. A genetic diagnosis has clinical utility as it may affect management and prognosis and informs recurrence risk, facilitating family planning decisions. More broadly, knowledge of disease genes increasingly allows population-based preconception carrier screening, which has reduced the incidence of recessive diseases in several populations. Despite gains in knowledge of the genetics of fetal akinesia, many families lack a genetic diagnosis. In this review, we describe the developments in Mendelian genetics of neuromuscular fetal akinesia in the genomics era. We examine genetic diagnoses with neuromuscular causes, specifically including the lower motor neuron, peripheral nerve, neuromuscular junction and muscle.
Collapse
Affiliation(s)
- Sarah Jane Beecroft
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QQ Block, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Marcus Lombard
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QQ Block, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - David Mowat
- Centre for Clinical Genetics, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | - Catriona McLean
- Victorian Neuromuscular Laboratory, Alfred Health, Melbourne, Victoria, Australia
| | - Anita Cairns
- Department of Neurology, Lady Cilento Children's Hospital, Brisbane, Queensland, Australia
| | - Mark Davis
- Neurogenetics Laboratory, Department of Diagnostic Genomics, PP Block, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Nigel G Laing
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QQ Block, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Gianina Ravenscroft
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia.,Harry Perkins Institute of Medical Research, QQ Block, QEII Medical Centre, Nedlands, Western Australia, Australia
| |
Collapse
|
49
|
Interactome analyses revealed that the U1 snRNP machinery overlaps extensively with the RNAP II machinery and contains multiple ALS/SMA-causative proteins. Sci Rep 2018; 8:8755. [PMID: 29884807 PMCID: PMC5993797 DOI: 10.1038/s41598-018-27136-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
Mutations in multiple RNA/DNA binding proteins cause Amyotrophic Lateral Sclerosis (ALS). Included among these are the three members of the FET family (FUS, EWSR1 and TAF15) and the structurally similar MATR3. Here, we characterized the interactomes of these four proteins, revealing that they largely have unique interactors, but share in common an association with U1 snRNP. The latter observation led us to analyze the interactome of the U1 snRNP machinery. Surprisingly, this analysis revealed the interactome contains ~220 components, and of these, >200 are shared with the RNA polymerase II (RNAP II) machinery. Among the shared components are multiple ALS and Spinal muscular Atrophy (SMA)-causative proteins and numerous discrete complexes, including the SMN complex, transcription factor complexes, and RNA processing complexes. Together, our data indicate that the RNAP II/U1 snRNP machinery functions in a wide variety of molecular pathways, and these pathways are candidates for playing roles in ALS/SMA pathogenesis.
Collapse
|
50
|
Burns DT, Donkervoort S, Müller JS, Knierim E, Bharucha-Goebel D, Faqeih EA, Bell SK, AlFaifi AY, Monies D, Millan F, Retterer K, Dyack S, MacKay S, Morales-Gonzalez S, Giunta M, Munro B, Hudson G, Scavina M, Baker L, Massini TC, Lek M, Hu Y, Ezzo D, AlKuraya FS, Kang PB, Griffin H, Foley AR, Schuelke M, Horvath R, Bönnemann CG. Variants in EXOSC9 Disrupt the RNA Exosome and Result in Cerebellar Atrophy with Spinal Motor Neuronopathy. Am J Hum Genet 2018; 102:858-873. [PMID: 29727687 PMCID: PMC5986733 DOI: 10.1016/j.ajhg.2018.03.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/06/2018] [Indexed: 12/30/2022] Open
Abstract
The exosome is a conserved multi-protein complex that is essential for correct RNA processing. Recessive variants in exosome components EXOSC3, EXOSC8, and RBM7 cause various constellations of pontocerebellar hypoplasia (PCH), spinal muscular atrophy (SMA), and central nervous system demyelination. Here, we report on four unrelated affected individuals with recessive variants in EXOSC9 and the effect of the variants on the function of the RNA exosome in vitro in affected individuals' fibroblasts and skeletal muscle and in vivo in zebrafish. The clinical presentation was severe, early-onset, progressive SMA-like motor neuronopathy, cerebellar atrophy, and in one affected individual, congenital fractures of the long bones. Three affected individuals of different ethnicity carried the homozygous c.41T>C (p.Leu14Pro) variant, whereas one affected individual was compound heterozygous for c.41T>C (p.Leu14Pro) and c.481C>T (p.Arg161∗). We detected reduced EXOSC9 in fibroblasts and skeletal muscle and observed a reduction of the whole multi-subunit exosome complex on blue-native polyacrylamide gel electrophoresis. RNA sequencing of fibroblasts and skeletal muscle detected significant >2-fold changes in genes involved in neuronal development and cerebellar and motor neuron degeneration, demonstrating the widespread effect of the variants. Morpholino oligonucleotide knockdown and CRISPR/Cas9-mediated mutagenesis of exosc9 in zebrafish recapitulated aspects of the human phenotype, as they have in other zebrafish models of exosomal disease. Specifically, portions of the cerebellum and hindbrain were absent, and motor neurons failed to develop and migrate properly. In summary, we show that variants in EXOSC9 result in a neurological syndrome combining cerebellar atrophy and spinal motoneuronopathy, thus expanding the list of human exosomopathies.
Collapse
|