1
|
Mohassel P, Hearn H, Rooney J, Zou Y, Johnson K, Norato G, Nalls MA, Yun P, Ogata T, Silverstein S, Sleboda DA, Roberts TJ, Rifkin DB, Bönnemann CG. Collagen type VI regulates TGF-β bioavailability in skeletal muscle in mice. J Clin Invest 2025; 135:e173354. [PMID: 40309777 PMCID: PMC12043086 DOI: 10.1172/jci173354] [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: 06/27/2023] [Accepted: 03/05/2025] [Indexed: 05/02/2025] Open
Abstract
Collagen VI-related disorders (COL6-RDs) are a group of rare muscular dystrophies caused by pathogenic variants in collagen VI genes (COL6A1, COL6A2, and COL6A3). Collagen type VI is a heterotrimeric, microfibrillar component of the muscle extracellular matrix (ECM), predominantly secreted by resident fibroadipogenic precursor cells in skeletal muscle. The absence or mislocalization of collagen VI in the ECM underlies the noncell-autonomous dysfunction and dystrophic changes in skeletal muscle with a yet elusive direct mechanistic link between the ECM and myofiber dysfunction. Here, we conducted a comprehensive natural history and outcome study in a mouse model of COL6-RDs (Col6a2-/- mice) using standardized (TREAT-NMD) functional, histological, and physiological parameters. Notably, we identify a conspicuous dysregulation of the TGF-β pathway early in the disease process and propose that the collagen VI-deficient matrix is not capable of regulating the dynamic TGF-β bioavailability both at baseline and in response to muscle injury. Thus, we propose a new mechanism for pathogenesis of the disease that links the ECM regulation of TGF-β with downstream skeletal muscle abnormalities, paving the way for the development and validation of therapeutics that target this pathway.
Collapse
Affiliation(s)
- Payam Mohassel
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hailey Hearn
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jachinta Rooney
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
| | - Yaqun Zou
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
| | - Kory Johnson
- Bioinformatics Section, Intramural Information Technology & Bioinformatics Program, National Institute of Neurological Disorders and Stroke, and
| | - Gina Norato
- Clinical Trials Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Matthew A. Nalls
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
| | - Pomi Yun
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
| | - Tracy Ogata
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
| | - Sarah Silverstein
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
| | - David A. Sleboda
- Department of Ecology and Evolutionary Biology, University of California, Irvine, Irvine, California, USA
| | - Thomas J. Roberts
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, USA
| | - Daniel B. Rifkin
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Carsten G. Bönnemann
- National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, NIH, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Barajaa MA, Ghosh D, Laurencin CT. Decellularized Extracellular Matrix-Derived Hydrogels: a Powerful Class of Biomaterials for Skeletal Muscle Regenerative Engineering Applications. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2025; 11:39-63. [PMID: 40201194 PMCID: PMC11978403 DOI: 10.1007/s40883-023-00328-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/18/2023] [Accepted: 11/28/2023] [Indexed: 04/10/2025]
Abstract
Purpose The extracellular matrix (ECM) is a complicated milieu consisting of structural and functional molecules secreted by the resident cells that provides an optimal microenvironmental niche for enhanced cell adhesion, growth, differentiation, and tissue formation and maturation. For decades, ECM bio-scaffolds prepared from decellularized tissues have been used to promote skeletal muscle regeneration; however, it was recently discovered that these decellularized ECM (dECM) materials can be further processed into hydrogels, thus expanding the potential applications of dECM materials in skeletal muscle regenerative engineerisng (SMRE). This review article highlights the recent advances in dECM-derived hydrogels toward skeletal muscle regeneration and repair. Method We screened articles in PubMed and bibliographic search using a combination of keywords. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Here, we discuss the skeletal muscle ECM's structure, function, and biochemical composition with emphasis on the role of the ECM during skeletal muscle embryogenesis, growth, development, and repair. Furthermore, we review various hydrogels used to promote skeletal muscle regeneration. We also review the current applications of dECM-derived hydrogels toward SMRE. Finally, we discuss the clinical translation potential of dECM-derived hydrogels for skeletal muscle regeneration and repair and their potential clinical considerations in the future. Conclusion Although much progress has been made in the field of dECM-derived hydrogels toward SMRE, it is still in its nascent stage. We believe improving and standardizing the methods of decellularization, lowering the immunogenicity of dECMs, and carrying out in vivo investigations in large animal models would advance their future clinical applications. Lay Summary Researchers have discovered an effective way to turn tissue materials into jelly-like substances known as extracellular matrix (ECM)-derived hydrogels. These ECM-derived hydrogels can help muscles heal better after serious injuries. They can be injected into gaps or used to guide muscle growth in the lab or body. This review article explains how these ECM-derived hydrogels are made and how they can be used to improve muscle healing. It also discusses their possible use in clinics and what needs to be considered before using them for medical treatments.
Collapse
Affiliation(s)
- Mohammed A. Barajaa
- Department of Biomedical Engineering, College of Engineering, Imam Abdulrahman Bin Faisal University, 34212 Dammam, Saudi Arabia
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical & Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
3
|
White T, López-Márquez A, Badosa C, Jimenez-Mallebrera C, Samitier J, Giannotti MI, Lagunas A. Nanomechanics of cell-derived matrices as a functional read-out in collagen VI-related congenital muscular dystrophies. J R Soc Interface 2025; 22:20240860. [PMID: 40070338 PMCID: PMC11897821 DOI: 10.1098/rsif.2024.0860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 03/15/2025] Open
Abstract
Changes in the mechanical properties of the extracellular matrix (ECM) are a hallmark of disease. Due to its relevance, several in vitro models have been developed for the ECM, including cell-derived matrices (CDMs). CDMs are decellularized natural ECMs assembled by cells that closely mimic the in vivo stromal fibre organization and molecular content. Here, we applied atomic force microscopy-force spectroscopy (AFM-FS) to evaluate the nanomechanical properties of CDMs obtained from patients diagnosed with collagen VI-related congenital muscular dystrophies (COL6-RDs). COL6-RDs are a set of neuromuscular conditions caused by pathogenic variants in any of the three major COL6 genes, which result in deficiency or dysfunction of the COL6 incorporated into the ECM of connective tissues. Current diagnosis includes the genetic confirmation of the disease and categorization of the phenotype based on maximum motor ability, as no direct correlation exists between genotype and phenotype of COL6-RDs. We describe differences in the elastic modulus (E) among CDMs from patients with different clinical phenotypes, as well as the restoration of E in CDMs obtained from genetically edited cells. Results anticipate the potential of the nanomechanical analysis of CDMs as a complementary clinical tool, providing phenotypic information about COL6-RDs and their response to gene therapies.
Collapse
Affiliation(s)
- Tom White
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Arístides López-Márquez
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Institut de Recerca Sant Joan de Déu, Barcelona, Catalunya, Spain
- Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Hospital Sant Joan de Déu, Barcelona, Catalunya, Spain
- CIBER-ER, ISCIII, Madrid, Spain
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Carmen Badosa
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Institut de Recerca Sant Joan de Déu, Barcelona, Catalunya, Spain
- Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Hospital Sant Joan de Déu, Barcelona, Catalunya, Spain
- CIBER-ER, ISCIII, Madrid, Spain
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Ceclila Jimenez-Mallebrera
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Institut de Recerca Sant Joan de Déu, Barcelona, Catalunya, Spain
- Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Hospital Sant Joan de Déu, Barcelona, Catalunya, Spain
- CIBER-ER, ISCIII, Madrid, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- CIBER-BBN, ISCIII, Madrid, Spain
- Department of Electronic and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Marina Inés Giannotti
- CIBER-BBN, ISCIII, Madrid, Spain
- Nanoprobes and Nanoswitches, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona, Spain
| | - Anna Lagunas
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- CIBER-BBN, ISCIII, Madrid, Spain
| |
Collapse
|
4
|
De Gregorio V, Barua M, Lennon R. Collagen formation, function and role in kidney disease. Nat Rev Nephrol 2025; 21:200-215. [PMID: 39548215 DOI: 10.1038/s41581-024-00902-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/17/2024]
Abstract
Highly abundant in mammals, collagens define the organization of tissues and participate in cell signalling. Most of the 28 vertebrate collagens, with the exception of collagens VI, VII, XXVI and XXVIII, can be categorized into five subgroups: fibrillar collagens, network-forming collagens, fibril-associated collagens with interrupted triple helices, membrane-associated collagens with interrupted triple helices and multiple triple-helix domains with interruptions. Collagen peptides are synthesized from the ribosome and enter the rough endoplasmic reticulum, where they undergo numerous post-translational modifications. The collagen chains form triple helices that can be secreted to form a diverse array of supramolecular structures in the extracellular matrix. Collagens are ubiquitously expressed and have been linked to a broad spectrum of disorders, including genetic disorders with kidney phenotypes. They also have an important role in kidney fibrosis and mass spectrometry-based proteomic studies have improved understanding of the composition of fibrosis in kidney disease. A wide range of therapeutics are in development for collagen and kidney disorders, including genetic approaches, chaperone therapies, protein degradation strategies and anti-fibrotic therapies. Improved understanding of collagens and their role in disease is needed to facilitate the development of more specific treatments for collagen and kidney disorders.
Collapse
Affiliation(s)
- Vanessa De Gregorio
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Moumita Barua
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada.
- Division of Nephrology, University Health Network, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester, UK.
| |
Collapse
|
5
|
McCarty RM, Saade D, Munot P, Laverty CG, Pinz H, Zou Y, McAnally M, Yun P, Tian C, Hu Y, Feng L, Phadke R, Ceulemans S, Magoulas P, Skalsky AJ, Friedman JR, Braddock SR, Neuhaus SB, Malicki DM, Bainbridge MN, Nahas S, Dimmock DP, Kingsmore SF, Lotze TE, Foley AR, Muntoni F, Straub V, Donkervoort S, Bönnemann CG. Clinical characterization of Collagen XII-related disease caused by biallelic COL12A1 variants. Ann Clin Transl Neurol 2025; 12:602-614. [PMID: 39923201 PMCID: PMC11920742 DOI: 10.1002/acn3.52225] [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: 05/21/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVE While there have been several reports of patients with dominantly acting COL12A1 variants, few cases of the more severe recessive Collagen XII-related disorders have previously been documented. METHODS We present detailed clinical, immunocytochemical, and imaging data on eight additional patients from seven families with biallelic pathogenic variants in COL12A1. RESULTS All patients presented with a consistent constellation of congenital onset clinical features: hypotonia, dysmorphic features, most notably gingival hypertrophy, prominent distal joint hyperlaxity, with co-occurring contractures of large joints, and variable muscle involvement, evident both clinically and on muscle imaging. Five patients presented with a severe congenital phenotype manifesting with profound weakness, significantly delayed or minimal attainment of motor milestones, respiratory insufficiency, and feeding difficulties. Three patients presented with mild-to-moderate muscle weakness and delayed milestones but were able to achieve independent ambulation. Patients were found to have biallelic loss-of-function COL12A1 variants, except for one family (p.I1393Ffs*11/p.A1110D). Consistent with the variable clinical spectrum, in vitro immunocytochemistry analysis in fibroblasts ranged from complete absence of Collagen XII expression in a patient with severe disease, to a mild reduction in a patient with milder disease. INTERPRETATION Here we characterize the clinical presentation, muscle imaging, and dermal fibroblast immunostaining findings associated with biallelic variants in COL12A1, further establishing COL12A1 as a recessive myopathic Ehlers-Danlos syndrome (mEDS) gene, and expanding the clinical spectrum to include a milder EDS phenotype.
Collapse
Affiliation(s)
- Riley M. McCarty
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Dimah Saade
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
- Department of Pediatrics, Roy J. and Lucille A. Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Pinki Munot
- The Dubowitz Neuromuscular CentreGreat Ormond Street Institute of Child HealthLondonUK
| | - Chamindra G. Laverty
- Department of NeurosciencesUniversity of California San DiegoSan DiegoCaliforniaUSA
| | - Hailey Pinz
- Division of Medical GeneticsSaint Louis UniversitySt. LouisMissouriUSA
| | - Yaqun Zou
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Meghan McAnally
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Pomi Yun
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Cuixia Tian
- Division of Neurology, Cincinnati Children's Hospital Medical Center & Department of PediatricsUniversity of CincinnatiCincinnatiOhioUSA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Lucy Feng
- The Dubowitz Neuromuscular CentreGreat Ormond Street Institute of Child HealthLondonUK
| | - Rahul Phadke
- Dubowitz Neuromuscular Centre, Division of NeuropathologyUCL Queen Square Institute of NeurologyQueen SquareLondonUK
| | - Sophia Ceulemans
- Division of GeneticsRady Children's HospitalSan DiegoCaliforniaUSA
| | - Pilar Magoulas
- Department of GeneticsTexas Children's HospitalHoustonTexasUSA
| | - Andrew J. Skalsky
- Division of Pediatric Rehabilitation MedicineRady's Children's HospitalSan DiegoCaliforniaUSA
- Department of Orthopaedic SurgeryUniversity of San DiegoSan DiegoCaliforniaUSA
| | - Jennifer R. Friedman
- Department of NeurosciencesUniversity of California San DiegoSan DiegoCaliforniaUSA
- Rady Children's Institute of Genomic MedicineSan DiegoCaliforniaUSA
- Department of PediatricsUniversity of California San DiegoSan DiegoCaliforniaUSA
| | | | - Sarah B. Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Denise M. Malicki
- Department of PathologyRady Children's HospitalSan DiegoCaliforniaUSA
| | | | - Shareef Nahas
- Rady Children's Institute of Genomic MedicineSan DiegoCaliforniaUSA
| | - David P. Dimmock
- Rady Children's Institute of Genomic MedicineSan DiegoCaliforniaUSA
| | | | - Timothy E. Lotze
- Division of Neurology and Developmental NeuroscienceBaylor College of Medicine/Texas Children's HospitalHoustonTexasUSA
| | - A. Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Francesco Muntoni
- The Dubowitz Neuromuscular CentreGreat Ormond Street Institute of Child HealthLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Volker Straub
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and StrokeNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
6
|
Aoki Y. Allele-specific CRISPR-Cas9 editing inactivates a single-nucleotide variant associated with collagen VI muscular dystrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102330. [PMID: 39380711 PMCID: PMC11460449 DOI: 10.1016/j.omtn.2024.102330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Affiliation(s)
- Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| |
Collapse
|
7
|
Efraim Y, Chen FYT, Niknezhad SV, Pham D, Cheong KN, An L, Sinada H, McNamara NA, Knox SM. Rebuilding the autoimmune-damaged corneal stroma through topical lubrication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.626078. [PMID: 39677756 PMCID: PMC11642755 DOI: 10.1101/2024.11.29.626078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Corneal lubrication is the most common treatment for relieving the signs and symptoms of dry eye and is considered to be largely palliative with no regenerative functions. Here we challenge this notion by demonstrating that wetting the desiccated cornea of an aqueous-deficient mouse model with the simplest form of lubrication, a saline-based solution, is sufficient to rescue the severely disrupted collagen-rich architecture of the stroma, the largest corneal compartment that is essential to transparency and vision. At the single cell level we show that stromal keratocytes responsible for maintaining stromal integrity are converted from an inflammatory state into unique reparative cell states by lubrication alone, thus revealing the extensive plasticity of these cells and the regenerative function of lubricating the surface. We further show that the generation of a reparative phenotype is due, in part, to disruption of an IL1β autocrine amplification loop promoting chronic inflammation. Thus, our study uncovers the regenerative potential of topical lubrication in dry eye and represents a paradigm shift in our understanding of its therapeutic impact.
Collapse
Affiliation(s)
- Yael Efraim
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Feeling Yu Ting Chen
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Seyyed Vahid Niknezhad
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Dylan Pham
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Ka Neng Cheong
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Luye An
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Hanan Sinada
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| | - Nancy A. McNamara
- School of Optometry and Vision Science Graduate Program, University of California, Berkeley; Oakland, CA 94720, USA
- Department of Anatomy, University of California, San Francisco; San Francisco, CA 94143, USA
| | - Sarah M. Knox
- Program in Craniofacial Biology, Department of Cell & Tissue Biology, University of California San Francisco; San Francisco, CA 94143, USA
| |
Collapse
|
8
|
Ceyhan AB, Kaynar A, Altay O, Zhang C, Temel SG, Turkez H, Mardinoglu A. Identifying Hub Genes and Metabolic Pathways in Collagen VI-Related Dystrophies: A Roadmap to Therapeutic Intervention. Biomolecules 2024; 14:1376. [PMID: 39595553 PMCID: PMC11592009 DOI: 10.3390/biom14111376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Collagen VI-related dystrophies (COL6RD) are a group of rare muscle disorders caused by mutations in specific genes responsible for type VI collagen production. It affects muscles, joints, and connective tissues, leading to weakness, joint problems, and structural issues. Currently, there is no effective treatment for COL6RD; its management typically addresses symptoms and complications. Therefore, it is essential to decipher the disease's molecular mechanisms, identify drug targets, and develop effective treatment strategies to treat COL6RD. In this study, we employed differential gene expression analysis, weighted gene co-expression network analysis, and genome-scale metabolic modeling to investigate gene expression patterns in COL6RD patients, uncovering key genes, significant metabolites, and disease-related pathophysiological pathways. First, we performed differential gene expression and weighted gene co-expression network analyses, which led to the identification of 12 genes (CHCHD10, MRPS24, TRIP10, RNF123, MRPS15, NDUFB4, COX10, FUNDC2, MDH2, RPL3L, NDUFB11, PARVB) as potential hub genes involved in the disease. Second, we utilized a drug repurposing strategy to identify pharmaceutical candidates that could potentially modulate these genes and be effective in the treatment. Next, we utilized context-specific genome-scale metabolic models to compare metabolic variations between healthy individuals and COL6RD patients. Finally, we conducted reporter metabolite analysis to identify reporter metabolites (e.g., phosphatidates, nicotinate ribonucleotide, ubiquinol, ferricytochrome C). In summary, our analysis revealed critical genes and pathways associated with COL6RD and identified potential targets, reporter metabolites, and candidate drugs for therapeutic interventions.
Collapse
Affiliation(s)
- Atakan Burak Ceyhan
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.B.C.); (A.K.)
| | - Ali Kaynar
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.B.C.); (A.K.)
| | - Ozlem Altay
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (O.A.); (C.Z.)
| | - Cheng Zhang
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (O.A.); (C.Z.)
| | - Sehime Gulsun Temel
- Department of Medical Genetics, Faculty of Medicine, Bursa Uludag University, Bursa 16059, Turkey;
- Department of Translational Medicine, Institute of Health Science, Bursa Uludag University, Bursa 16059, Turkey
- Department of Histology and Embryology, Faculty of Medicine, Bursa Uludag University, Bursa 16059, Turkey
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum 25030, Turkey;
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (A.B.C.); (A.K.)
- Science for Life Laboratory, KTH—Royal Institute of Technology, SE-17165 Stockholm, Sweden; (O.A.); (C.Z.)
| |
Collapse
|
9
|
Liang Z, Huang T, Li W, Ma Z, Wang K, Zhai Z, Fan Y, Fu Y, Wang X, Qin Y, Wang B, Zhao C, Kuang J, Pei D. ALKBH5 governs human endoderm fate by regulating the DKK1/4-mediated Wnt/β-catenin activation. Nucleic Acids Res 2024; 52:10879-10896. [PMID: 39166492 PMCID: PMC11472173 DOI: 10.1093/nar/gkae707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/25/2024] [Accepted: 08/03/2024] [Indexed: 08/23/2024] Open
Abstract
N6-methyladenonsine (m6A) is ubiquitously distributed in mammalian mRNA. However, the precise involvement of m6A in early development has yet to be fully elucidated. Here, we report that deletion of the m6A demethylase ALKBH5 in human embryonic stem cells (hESCs) severely impairs definitive endoderm (DE) differentiation. ALKBH5-/- hESCs fail to undergo the primitive streak (PS) intermediate transition that precedes endoderm specification. Mechanistically, we show that ALKBH5 deficiency induces m6A hypermethylation around the 3' untranslated region (3'UTR) of GATA6 transcripts and destabilizes GATA6 mRNA in a YTHDF2-dependent manner. Moreover, GATA6 binds to the promoters of critical regulatory genes involved in Wnt/β-catenin signaling transduction, including the canonical Wnt antagonist DKK1 and DKK4, which are unexpectedly repressed upon the dysregulation of GATA6 mRNA metabolism. Remarkably, DKK1 and DKK4 both exhibit a pleiotropic effect in modulating the Wnt/β-catenin cascade and guard the endogenous signaling activation underlying DE formation as potential downstream targets of the ALKBH5-GATA6 regulation. Here, we unravel a role of ALKBH5 in human endoderm formation in vitro by modulating the canonical Wnt signaling logic through the previously unrecognized functions of DKK1/4, thus capturing a more comprehensive role of m6A in early human embryogenesis.
Collapse
Affiliation(s)
- Zechuan Liang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Tao Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Wei Li
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhaoyi Ma
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Kaipeng Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Fudan Unversity, Shanghai, China
| | - Ziwei Zhai
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yixin Fan
- CAS Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Fu
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Fudan Unversity, Shanghai, China
| | - Xiaomin Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Yue Qin
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Bo Wang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang University of Science and Technology School of Information and Electronic Engineering, Hangzhou, China
- Zhejiang Key Laboratory of Biomedical Intelligent Computing Technology, Hangzhou, China
| | - Chengchen Zhao
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Zhejiang Key Laboratory of Biomedical Intelligent Computing Technology, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
| | - Junqi Kuang
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
| |
Collapse
|
10
|
Sharaf-Eldin WE, Rafat K, Issa MY, Elbendary HM, Eissa NR, Hawaary B, Gaboon NEA, Maroofian R, Gleeson JG, Essawi ML, Zaki MS. Clinical and Molecular Profiles of a Cohort of Egyptian Patients with Collagen VI-Related Dystrophy. J Mol Neurosci 2024; 74:93. [PMID: 39367186 PMCID: PMC11452470 DOI: 10.1007/s12031-024-02266-8] [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: 06/30/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024]
Abstract
Collagen VI-related dystrophies (COL6-RD) display a wide spectrum of disease severity and genetic variability ranging from mild Bethlem myopathy (BM) to severe Ullrich congenital muscular dystrophy (UCMD) and the intermediate severities in between with dual modes of inheritance, dominant and recessive. In the current study, next-generation sequencing demonstrated potential variants in the genes coding for the three alpha chains of collagen VI (COL6A1, COL6A2, or COL6A3) in a cohort of Egyptian patients with progressive muscle weakness (n = 23). Based on the age of disease onset and the patient clinical course, subjects were diagnosed as follows: 12 with UCMD, 8 with BM, and 3 with intermediate disease form. Fourteen pathogenic variants, including 5 novel alterations, were reported in the enrolled subjects. They included 3 missense, 3 frameshift, and 6 splicing variants in 4, 3, and 6 families, respectively. In addition, a nonsense variant in a single family and an inframe variant in 3 different families were also detected. Recessive and dominant modes of inheritance were recorded in 9 and 8 families, respectively. According to ACMG guidelines, variants were classified as pathogenic (n = 7), likely pathogenic (n = 4), or VUS (n = 3) with significant pathogenic potential. To our knowledge, the study provided the first report of the clinical and genetic findings of a cohort of Egyptian patients with collagen VI deficiency. Inter- and intra-familial clinical variability was evident among the study cohort.
Collapse
Affiliation(s)
- Wessam E Sharaf-Eldin
- Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Karima Rafat
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt
| | - Mahmoud Y Issa
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt
| | - Hasnaa M Elbendary
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt
| | - Noura R Eissa
- Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Bahaa Hawaary
- Pediatrics Department, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Nagwa E A Gaboon
- Medical Genetics Centre, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Medical Genetics Department, Armed Forces College of Medicine, Cairo, Egypt
| | - Reza Maroofian
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, UK
| | - Joseph G Gleeson
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093, USA
- Rady Children's Institute for Genomic Medicine, San Diego, La Jolla, CA, 92093, USA
| | - Mona L Essawi
- Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, 12311, Egypt.
- Medical Genetics Department, Armed Forces College of Medicine, Cairo, Egypt.
| |
Collapse
|
11
|
Gai X, Xia Q, Wang H, Bi H, Wang J, Zhao Y. Study on the mechanism of echinacoside in preventing and treating hypoxic pulmonary hypertension based on proteomic analyses. Pharmacol Res Perspect 2024; 12:e70025. [PMID: 39401152 PMCID: PMC11472809 DOI: 10.1002/prp2.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/17/2024] Open
Abstract
Hypoxic pulmonary hypertension (HPH), a chronic condition affecting the cardiopulmonary system, has high mortality. Echinacoside (ECH) is a phenylethanoid glycoside, which is used to ameliorate pulmonary vascular remodeling and pulmonary vasoconstriction in rats. Accordingly, we aimed to explore the mechanism of ECH in preventing and treating HPH. Sprague Dawley rats were housed in a hypobaric hypoxia chamber for 28 days to obtain the HPH model. The experimental rats were randomly allocated into the following several groups: normoxia group, chronic hypoxia group, and ECH group. The therapeutic results of ECH (10, 20, and 40 mg/kg) showed that ECH reduced mPAP, Hb, Hct, and RVHI in HPH rats. Then this work employed label-free quantitative proteomic analysis, western blotting, and RT-PCR to investigate the mechanism by which ECH prevents HPH. The results found that in the chronic hypoxia group, the levels of ACSL1, COL6A1, COL4A2, COL1A1, and PC increased compared to the normoxia group. However, the opposite effect was observed in the chronic hypoxia group treated with ECH. The study indicates that the administration of ECH may slow the pathological progression of HPH by suppressing the inflammatory response, inhibiting smooth muscle cell proliferation, and minimizing the deposition of extracellular matrix.
Collapse
Affiliation(s)
- Xiangyun Gai
- Department of PharmacyQinghai Minzu UniversityXiningChina
| | - Qingqing Xia
- Department of PharmacyQinghai Minzu UniversityXiningChina
| | - Hongmai Wang
- Department of PharmacyQinghai Minzu UniversityXiningChina
| | - Hongtao Bi
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety EvaluationNorthwest Institute of Plateau Biology, Chinese Academy of ScienceXiningChina
| | - Jinyu Wang
- Department of PharmacyQinghai Minzu UniversityXiningChina
| | - Yuefu Zhao
- Department of PharmacyQinghai Minzu UniversityXiningChina
| |
Collapse
|
12
|
Findlay AR. Dominantly inherited muscle disorders: understanding their complexity and exploring therapeutic approaches. Dis Model Mech 2024; 17:dmm050720. [PMID: 39501809 PMCID: PMC11574355 DOI: 10.1242/dmm.050720] [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] [Indexed: 11/21/2024] Open
Abstract
Treatments for disabling and life-threatening hereditary muscle disorders are finally close to becoming a reality. Research has thus far focused primarily on recessive forms of muscle disease. The gene replacement strategies that are commonly employed for recessive, loss-of-function disorders are not readily translatable to most dominant myopathies owing to the presence of a normal chromosome in each nucleus, hindering the development of novel treatments for these dominant disorders. This is largely due to their complex, heterogeneous disease mechanisms that require unique therapeutic approaches. However, as viral and RNA interference-based therapies enter clinical use, key tools are now in place to develop treatments for dominantly inherited disorders of muscle. This article will review what is known about dominantly inherited disorders of muscle, specifically their genetic basis, how mutations lead to disease, and the pathomechanistic implications for therapeutic approaches.
Collapse
Affiliation(s)
- Andrew R Findlay
- Washington University Saint Louis, Neuromuscular Disease Center, 660 S. Euclid Ave., St Louis, MO 63110, USA
| |
Collapse
|
13
|
Bolduc V, Sizov K, Brull A, Esposito E, Chen GS, Uapinyoying P, Sarathy A, Johnson KR, Bönnemann CG. Allele-specific CRISPR-Cas9 editing inactivates a single nucleotide variant associated with collagen VI muscular dystrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102269. [PMID: 39171142 PMCID: PMC11338111 DOI: 10.1016/j.omtn.2024.102269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/12/2024] [Indexed: 08/23/2024]
Abstract
The application of allele-specific gene editing tools can expand the therapeutic options for dominant genetic conditions, either via gene correction or via allelic gene inactivation in situations where haploinsufficiency is tolerated. Here, we used allele-targeted CRISPR-Cas9 guide RNAs (gRNAs) to introduce inactivating frameshifting indels at an SNV in the COL6A1 gene (c.868G>A; G290R), a variant that acts as dominant negative and that is associated with a severe form of congenital muscular dystrophy. We expressed SpCas9 along with allele-targeted gRNAs, without providing a repair template, in primary fibroblasts derived from four patients and one control subject. Amplicon deep sequencing for two gRNAs tested showed that single-nucleotide deletions accounted for the majority of indels introduced. While activity of the two gRNAs was greater at the G290R allele, both gRNAs were also active at the wild-type allele. To enhance allele selectivity, we introduced deliberate additional mismatches to one gRNA. One of these optimized gRNAs showed minimal activity at the WT allele, while generating productive edits and improving collagen VI matrix in cultured patient fibroblasts. This study strengthens the potential of gene editing to treat dominant-negative disorders, but also underscores the challenges in achieving allele selectivity with gRNAs.
Collapse
Affiliation(s)
- Véronique Bolduc
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine Sizov
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Astrid Brull
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eric Esposito
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Grace S. Chen
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Prech Uapinyoying
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Center for Genetic Medicine Research, Children’s National Research and Innovation Campus, Children’s National Hospital, Washington, DC 20012, USA
| | - Apurva Sarathy
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kory R. Johnson
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carsten G. Bönnemann
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
İpek R, Çavdartepe BE, Bozdoğan ST, Yiş U. COL12A1 Gene Variant and a Review of the Literature: A Case Report of Ullrich Congenital Muscular Dystrophy. Mol Syndromol 2024; 15:311-316. [PMID: 39129837 PMCID: PMC11316441 DOI: 10.1159/000536344] [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: 06/29/2023] [Accepted: 01/16/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Mutations in collagen type IV-associated genes lead to Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy (BM). COL12A1 gene mutations have rarely been reported in patients with UCMD- and BM-like disorders not involving COL6 mutations. UCMD-2 results from homozygous mutations in the COL12A1 gene on the long arm of chromosome 6. Pathogenic variants in COL12A1 result in a rare congenital connective tissue/myopathy overlap syndrome under the heading of myopathic Ehlers-Danlos syndrome. COL12A1 dominant pathogenic variants have been rarely reported, and the phenotypic spectrum has not yet been identified. Case Presentation We describe a female patient aged 2 years and 10 months exhibiting a milder phenotype who presented due to pronounced joint hyperlaxity, frequent falls, and skin lesions. Genetic analysis revealed a homozygous c.8903C>T (p.Pro2968Leu) missense variant that had previously been described but concerning which there had been no clinical report, in the COL12A1 gene. Discussion/Conclusion This report is presented in order to raise awareness of rare mutations in the COL12A1 gene that affect muscle and connective tissue and to add to the literature in defining the phenotypic spectrum.
Collapse
Affiliation(s)
- Rojan İpek
- Department of Pediatric Neurology, Dicle University, Diyarbakır, Turkey
| | | | | | - Uluç Yiş
- Departmant of Pediatric Neurology, Dokuz Eylül University, İzmir, Turkey
| |
Collapse
|
15
|
Kachuei M, Orangi K, Mohammadi A, Mohammadi M, Mojbafan M. Bethlem myopathy: A novel homozygous variant of c.385C>T (p.Arg129Cys) in the COL6A2 gene. Clin Case Rep 2024; 12:e9306. [PMID: 39135765 PMCID: PMC11317720 DOI: 10.1002/ccr3.9306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Key Clinical Message This case highlights the challenges in diagnosing Bethlem myopathy, the need for a high index of suspicion, and the importance of recognizing the diverse clinical presentations of this rare condition. Enhanced understanding can aid in early diagnosis and tailored management. Abstract Bethlem myopathy (BM), a rare collagen VI-related myopathy, is characterized by progressive muscle weakness and contractures, typically affecting the proximal muscles and joints. This case report presents a 15-year-old girl from Tehran, Iran, with a 5-year history of severe limb pain and progressive weakness. Born to consanguineous parents, the patient displayed delayed walking milestones and significant hypotonia, leading to a waddling gait and lumbar hyperlordosis. Neurological examination revealed marked proximal lower limb weakness, a positive Gowers' sign, and absent myotatic reflexes. Elevated creatine phosphokinase (CPK) levels and electromyography (EMG) results indicated myopathy, while nerve conduction studies showed no neuropathy. Genetic testing revealed a novel homozygous variant of c.385C>T (p.Arg129Cys) in the COL6A2 gene, classified as a variant of uncertain significance (VUS) per American College of Medical Genetics and Genomics (ACMG) guidelines due to its rarity and specific phenotype association. Differential diagnosis is essential to distinguish it from other neuromuscular conditions. Management primarily focuses on symptom relief and enhancing patients' quality of life. This case highlights the challenges in diagnosing BM, the need for a high index of suspicion, and the importance of recognizing the diverse clinical presentations of this rare condition. Enhanced understanding can aid in early diagnosis and tailored management.
Collapse
Affiliation(s)
- Maryam Kachuei
- Firoozabadi Clinical Research Development Unit, Department of PediatricsIran University of Medical SciencesTehranIran
| | - Kiana Orangi
- School of MedicineIran University of Medical SciencesTehranIran
| | - Aynaz Mohammadi
- School of MedicineIran University of Medical SciencesTehranIran
| | | | - Marzieh Mojbafan
- Department of Medical Genetics, School of MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
16
|
Brull A, Sarathy A, Bolduc V, Chen GS, McCarty RM, Bönnemann CG. Optimized allele-specific silencing of the dominant-negative COL6A1 G293R substitution causing collagen VI-related dystrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102178. [PMID: 38617974 PMCID: PMC11015156 DOI: 10.1016/j.omtn.2024.102178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
Collagen VI-related dystrophies (COL6-RDs) are a group of severe, congenital-onset muscular dystrophies for which there is no effective causative treatment. Dominant-negative mutations are common in COL6A1, COL6A2, and COL6A3 genes, encoding the collagen α1, α2, and α3 (VI) chains. They act by incorporating into the hierarchical assembly of the three α (VI) chains and consequently produce a dysfunctional collagen VI extracellular matrix, while haploinsufficiency for any of the COL6 genes is not associated with disease. Hence, allele-specific transcript inactivation is a valid therapeutic strategy, although selectively targeting a pathogenic single nucleotide variant is challenging. Here, we develop a small interfering RNA (siRNA) that robustly, and in an allele-specific manner, silences a common glycine substitution (G293R) caused by a single nucleotide change in COL6A1 gene. By intentionally introducing an additional mismatch into the siRNA design, we achieved enhanced specificity toward the mutant allele. Treatment of patient-derived fibroblasts effectively reduced the levels of mutant transcripts while maintaining unaltered wild-type transcript levels, rescuing the secretion and assembly of collagen VI matrix by reducing the dominant-negative effect of mutant chains. Our findings establish a promising treatment approach for patients with the recurrent dominantly negative acting G293R glycine substitution.
Collapse
Affiliation(s)
- Astrid Brull
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Apurva Sarathy
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Véronique Bolduc
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Grace S. Chen
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Riley M. McCarty
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carsten G. Bönnemann
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Chen WQ, Yuan YF, Hu KN, Sun DL, Wang SW, He QB, Liu YM, Han CY, Zhang J, Li YZ. Identification of novel variations in three cases with rare inherited neuromuscular disorder. Exp Ther Med 2024; 27:270. [PMID: 38756899 PMCID: PMC11097291 DOI: 10.3892/etm.2024.12558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/22/2024] [Indexed: 05/18/2024] Open
Abstract
Inherited neuromuscular disorder (IND) is a broad-spectrum, clinically diverse group of diseases that are caused due to defects in the neurosystem, muscles and related tissue. Since IND may originate from mutations in hundreds of different genes, the resulting heterogeneity of IND is a great challenge for accurate diagnosis and subsequent management. Three pediatric cases with IND were enrolled in the present study and subjected to a thorough clinical examination. Next, a genetic investigation was conducted using whole-exome sequencing (WES). The suspected variants were validated through Sanger sequencing or quantitative fluorescence PCR assay. A new missense variant of the Spastin (SPAST) gene was found and analyzed at the structural level using molecular dynamics (MD) simulations. All three cases presented with respective specific clinical manifestations, which reflected the diversity of IND. WES detected the diagnostic variants in all 3 cases: A compound variation comprising collagen type VI α3 chain (COL6A3) (NM_004369; exon19):c.6322G>T(p.E1208*) and a one-copy loss of COL6A3:exon19 in Case 1, which are being reported for the first time; a de novo SPAST (NM_014946; exon8):c.1166C>A(p.T389K) variant in Case 2; and a de novo Duchenne muscular dystrophy (NM_004006; exon11):c.1150-17_1160delACTTCCTTCTTTGTCAGGGGTACATGATinsC variant in Case 3. The structural and MD analyses revealed that the detected novel SPAST: c.1166C>A(p.T389K) variant mainly altered the intramolecular hydrogen bonding status and the protein segment's secondary structure. In conclusion, the present study expanded the IND mutation spectrum. The study not only detailed the precise diagnoses of these cases but also furnished substantial grounds for informed consultations. The approach involving the genetic evaluation strategy using WES for variation screening followed by validation using appropriate methods is beneficial due to the considerable heterogeneity of IND.
Collapse
Affiliation(s)
- Wen-Qi Chen
- Prenatal Diagnosis Center, Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei 050011, P.R. China
- Hebei Key Laboratory of Maternal and Fetal Medicine, Shijiazhuang, Hebei 050011, P.R. China
- Shijiazhuang Key Laboratory of Reproductive Health, Shijiazhuang, Hebei 050011, P.R. China
| | - Yu-Fan Yuan
- Hebei Key Laboratory of Maternal and Fetal Medicine, Shijiazhuang, Hebei 050011, P.R. China
- Shijiazhuang Key Laboratory of Reproductive Health, Shijiazhuang, Hebei 050011, P.R. China
| | - Ke-Na Hu
- Hebei Key Laboratory of Maternal and Fetal Medicine, Shijiazhuang, Hebei 050011, P.R. China
- Shijiazhuang Key Laboratory of Reproductive Health, Shijiazhuang, Hebei 050011, P.R. China
| | - Dong-Lan Sun
- Prenatal Diagnosis Center, Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei 050011, P.R. China
- Hebei Key Laboratory of Maternal and Fetal Medicine, Shijiazhuang, Hebei 050011, P.R. China
- Shijiazhuang Key Laboratory of Reproductive Health, Shijiazhuang, Hebei 050011, P.R. China
| | - Si-Wen Wang
- Prenatal Diagnosis Center, Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei 050011, P.R. China
- Hebei Key Laboratory of Maternal and Fetal Medicine, Shijiazhuang, Hebei 050011, P.R. China
- Shijiazhuang Key Laboratory of Reproductive Health, Shijiazhuang, Hebei 050011, P.R. China
| | - Qing-Bing He
- Department of Pediatric Orthopaedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yan-Ming Liu
- Prenatal Diagnosis Center, Langfang Maternal and Child Health Care Hospital, Langfang, Hebei 065000, P.R. China
| | - Cong-Ying Han
- Prenatal Diagnosis Center, Langfang Maternal and Child Health Care Hospital, Langfang, Hebei 065000, P.R. China
| | - Jing Zhang
- Prenatal Diagnosis Center, Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei 050011, P.R. China
- Hebei Key Laboratory of Maternal and Fetal Medicine, Shijiazhuang, Hebei 050011, P.R. China
- Shijiazhuang Key Laboratory of Reproductive Health, Shijiazhuang, Hebei 050011, P.R. China
| | - Ya-Zhou Li
- Department of Pediatric Orthopaedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
18
|
Wilpert NM, Schuelke M, Lala B, Weiß C. A Mild But Typical Presentation of Bethlem Myopathy With a Novel In-Frame Deletion in COL6A1: Almost Overlooked. Neurology 2024; 102:e209476. [PMID: 38754040 DOI: 10.1212/wnl.0000000000209476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Affiliation(s)
- Nina-Maria Wilpert
- From the Department of Neuropediatrics (N.-M.W., M.S., C.W.); Center for Chronically Sick Children (N.-M.W., M.S., C.W.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin; BIH Charité Junior Clinician Scientist Program (N.-M.W.), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy; and Department of Pediatric Radiology (B.L.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Germany
| | - Markus Schuelke
- From the Department of Neuropediatrics (N.-M.W., M.S., C.W.); Center for Chronically Sick Children (N.-M.W., M.S., C.W.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin; BIH Charité Junior Clinician Scientist Program (N.-M.W.), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy; and Department of Pediatric Radiology (B.L.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Germany
| | - Birgit Lala
- From the Department of Neuropediatrics (N.-M.W., M.S., C.W.); Center for Chronically Sick Children (N.-M.W., M.S., C.W.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin; BIH Charité Junior Clinician Scientist Program (N.-M.W.), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy; and Department of Pediatric Radiology (B.L.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Germany
| | - Claudia Weiß
- From the Department of Neuropediatrics (N.-M.W., M.S., C.W.); Center for Chronically Sick Children (N.-M.W., M.S., C.W.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin; BIH Charité Junior Clinician Scientist Program (N.-M.W.), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy; and Department of Pediatric Radiology (B.L.), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Germany
| |
Collapse
|
19
|
McGarrigle C, McGrath L, Khan E. Anaesthesia management of a patient with Bethlem Myopathy for elective tonsillectomy: a case report. BMC Anesthesiol 2024; 24:173. [PMID: 38730355 PMCID: PMC11083902 DOI: 10.1186/s12871-024-02539-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Bethlem Myopathy is a collagen VI-related myopathy presenting as a rare hereditary muscular disorder with progressive muscular weakness and joint contractures. Despite its milder clinical course relative to other myopathies, anaesthetic management can be challenging. High arched palates and fixed flexion deformities may contribute to a difficult airway. A progressive decline in pulmonary function can present later into adulthood. This respiratory decline can carry secondary cardiovascular consequences due to the progressive nature of restrictive lung disease, including right sided heart disease and pulmonary hypertension. We describe a case of a male patient with Bethlem Myopathy undergoing anaesthesia, to contribute to the limited body of literature on this condition and enhance awareness and guidance amongst anaesthesiologists on approaching patients with this condition. This is the first case report within the literature of its kind. CASE PRESENTATION This case details a 33-year-old male with Bethlem Myopathy undergoing tonsillectomy. Diagnosed in childhood following developmental delays, the patient had no prior anaesthetic exposure and no family history of anaesthetic complications. Anaesthetic induction was achieved without complications, avoiding depolarizing muscle relaxants and careful airway management. Extreme care was taken in patient positioning to prevent complications. The surgery proceeded without incident and muscle paralysis was reversed with Suggammadex, resulting in no adverse post-operative respiratory complications. The patient was discharged on the first post-operative day without any respiratory or cardiovascular compromise. CONCLUSIONS Bethlem Myopathy, while often exhibiting a mild clinical course, can present anaesthetic challenges. Awareness of potential complications including a difficult airway, cardiovascular and respiratory implications as well as the need for specialised monitoring and positioning is crucial to ensure a safe peri-operative course.
Collapse
|
20
|
Ducommun S, Jannig PR, Cervenka I, Murgia M, Mittenbühler MJ, Chernogubova E, Dias JM, Jude B, Correia JC, Van Vranken JG, Ocana-Santero G, Porsmyr-Palmertz M, McCann Haworth S, Martínez-Redondo V, Liu Z, Carlström M, Mann M, Lanner JT, Teixeira AI, Maegdefessel L, Spiegelman BM, Ruas JL. Mustn1 is a smooth muscle cell-secreted microprotein that modulates skeletal muscle extracellular matrix composition. Mol Metab 2024; 82:101912. [PMID: 38458566 PMCID: PMC10950823 DOI: 10.1016/j.molmet.2024.101912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024] Open
Abstract
OBJECTIVE Skeletal muscle plasticity and remodeling are critical for adapting tissue function to use, disuse, and regeneration. The aim of this study was to identify genes and molecular pathways that regulate the transition from atrophy to compensatory hypertrophy or recovery from injury. Here, we have used a mouse model of hindlimb unloading and reloading, which causes skeletal muscle atrophy, and compensatory regeneration and hypertrophy, respectively. METHODS We analyzed mouse skeletal muscle at the transition from hindlimb unloading to reloading for changes in transcriptome and extracellular fluid proteome. We then used qRT-PCR, immunohistochemistry, and bulk and single-cell RNA sequencing data to determine Mustn1 gene and protein expression, including changes in gene expression in mouse and human skeletal muscle with different challenges such as exercise and muscle injury. We generated Mustn1-deficient genetic mouse models and characterized them in vivo and ex vivo with regard to muscle function and whole-body metabolism. We isolated smooth muscle cells and functionally characterized them, and performed transcriptomics and proteomics analysis of skeletal muscle and aorta of Mustn1-deficient mice. RESULTS We show that Mustn1 (Musculoskeletal embryonic nuclear protein 1, also known as Mustang) is highly expressed in skeletal muscle during the early stages of hindlimb reloading. Mustn1 expression is transiently elevated in mouse and human skeletal muscle in response to intense exercise, resistance exercise, or injury. We find that Mustn1 expression is highest in smooth muscle-rich tissues, followed by skeletal muscle fibers. Muscle from heterozygous Mustn1-deficient mice exhibit differences in gene expression related to extracellular matrix and cell adhesion, compared to wild-type littermates. Mustn1-deficient mice have normal muscle and aorta function and whole-body glucose metabolism. We show that Mustn1 is secreted from smooth muscle cells, and that it is present in arterioles of the muscle microvasculature and in muscle extracellular fluid, particularly during the hindlimb reloading phase. Proteomics analysis of muscle from Mustn1-deficient mice confirms differences in extracellular matrix composition, and female mice display higher collagen content after chemically induced muscle injury compared to wild-type littermates. CONCLUSIONS We show that, in addition to its previously reported intracellular localization, Mustn1 is a microprotein secreted from smooth muscle cells into the muscle extracellular space. We explore its role in muscle ECM deposition and remodeling in homeostasis and upon muscle injury. The role of Mustn1 in fibrosis and immune infiltration upon muscle injury and dystrophies remains to be investigated, as does its potential for therapeutic interventions.
Collapse
Affiliation(s)
- Serge Ducommun
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Paulo R Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Igor Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Marta Murgia
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi, 58/B, 35131 Padua, Italy; Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Melanie J Mittenbühler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ekaterina Chernogubova
- Department of Medicine, Cardiovascular Unit, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - José M Dias
- Department of Cell and Molecular Biology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden; Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Baptiste Jude
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jorge C Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Gabriel Ocana-Santero
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Margareta Porsmyr-Palmertz
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sarah McCann Haworth
- Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Vicente Martínez-Redondo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Zhengye Liu
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Matthias Mann
- Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Johanna T Lanner
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ana I Teixeira
- Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Lars Maegdefessel
- Department of Medicine, Cardiovascular Unit, Karolinska Institutet, 171 77 Stockholm, Sweden; Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; German Center for Cardiovascular Research DZHK, Partner Site Munich Heart Alliance, 10785 Berlin, Germany
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jorge L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Pharmacology and Stanley and Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
21
|
Maksiutenko EM, Merkureva V, Barbitoff YA, Tsay VV, Aseev MV, Glotov AS, Glotov OS. Exome sequencing in extreme altitude mountaineers identifies pathogenic variants in RTEL1 and COL6A1 previously associated with respiratory failure. Physiol Rep 2024; 12:e16015. [PMID: 38653581 PMCID: PMC11039409 DOI: 10.14814/phy2.16015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Adaptation of humans to challenging environmental conditions, such as extreme temperature, malnutrition, or hypoxia, is an interesting phenomenon for both basic and applied research. Identification of the genetic factors contributing to human adaptation to these conditions enhances our understanding of the underlying molecular and physiological mechanisms. In our study, we analyzed the exomes of 22 high altitude mountaineers to uncover genetic variants contributing to hypoxic adaptation. To our surprise, we identified two putative loss-of-function variants, rs1385101139 in RTEL1 and rs1002726737 in COL6A1 in two extremely high altitude (personal record of more than 8500 m) professional climbers. Both variants can be interpreted as pathogenic according to medical geneticists' guidelines, and are linked to inherited conditions involving respiratory failure (late-onset pulmonary fibrosis and severe Ullrich muscular dystrophy for rs1385101139 and rs1002726737, respectively). Our results suggest that a loss of gene function may act as an important factor of human adaptation, which is corroborated by previous reports in other human subjects.
Collapse
Affiliation(s)
- Evgeniia M. Maksiutenko
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
| | - Valeriia Merkureva
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- CerbaLab Ltd.St. PetersburgRussia
| | - Yury A. Barbitoff
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- Department of Genetics and BiotechnologySt. Petersburg State UniversitySt. PetersburgRussia
| | - Victoria V. Tsay
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- CerbaLab Ltd.St. PetersburgRussia
- Department of Experimental Medical VirologyMolecular Genetics and Biobanking of Pediatric Research and Clinical Center for Infectious DiseasesSt. PetersburgRussia
| | - Mikhail V. Aseev
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- CerbaLab Ltd.St. PetersburgRussia
| | - Andrey S. Glotov
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- Department of Genetics and BiotechnologySt. Petersburg State UniversitySt. PetersburgRussia
| | - Oleg S. Glotov
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- Department of Experimental Medical VirologyMolecular Genetics and Biobanking of Pediatric Research and Clinical Center for Infectious DiseasesSt. PetersburgRussia
| |
Collapse
|
22
|
Foley AR, Bolduc V, Guirguis F, Donkervoort S, Hu Y, Orbach R, McCarty RM, Sarathy A, Norato G, Cummings BB, Lek M, Sarkozy A, Butterfield RJ, Kirschner J, Nascimento A, Benito DND, Quijano-Roy S, Stojkovic T, Merlini L, Comi G, Ryan M, McDonald D, Munot P, Yoon G, Leung E, Finanger E, Leach ME, Collins J, Tian C, Mohassel P, Neuhaus SB, Saade D, Cocanougher BT, Chu ML, Scavina M, Grosmann C, Richardson R, Kossak BD, Gospe SM, Bhise V, Taurina G, Lace B, Troncoso M, Shohat M, Shalata A, Chan SH, Jokela M, Palmio J, Haliloğlu G, Jou C, Gartioux C, Solomon-Degefa H, Freiburg CD, Schiavinato A, Zhou H, Aguti S, Nevo Y, Nishino I, Jimenez-Mallebrera C, Lamandé SR, Allamand V, Gualandi F, Ferlini A, MacArthur DG, Wilton SD, Wagener R, Bertini E, Muntoni F, Bönnemann CG. The recurrent deep intronic pseudoexon-inducing variant COL6A1 c.930+189C>T results in a consistently severe phenotype of COL6-related dystrophy: Towards clinical trial readiness for splice-modulating therapy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.29.24304673. [PMID: 38585825 PMCID: PMC10996746 DOI: 10.1101/2024.03.29.24304673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Collagen VI-related dystrophies (COL6-RDs) manifest with a spectrum of clinical phenotypes, ranging from Ullrich congenital muscular dystrophy (UCMD), presenting with prominent congenital symptoms and characterised by progressive muscle weakness, joint contractures and respiratory insufficiency, to Bethlem muscular dystrophy, with milder symptoms typically recognised later and at times resembling a limb girdle muscular dystrophy, and intermediate phenotypes falling between UCMD and Bethlem muscular dystrophy. Despite clinical and immunohistochemical features highly suggestive of COL6-RD, some patients had remained without an identified causative variant in COL6A1, COL6A2 or COL6A3. With combined muscle RNA-sequencing and whole-genome sequencing we uncovered a recurrent, de novo deep intronic variant in intron 11 of COL6A1 (c.930+189C>T) that leads to a dominantly acting in-frame pseudoexon insertion. We subsequently identified and have characterised an international cohort of forty-four patients with this COL6A1 intron 11 causative variant, one of the most common recurrent causative variants in the collagen VI genes. Patients manifest a consistently severe phenotype characterised by a paucity of early symptoms followed by an accelerated progression to a severe form of UCMD, except for one patient with somatic mosaicism for this COL6A1 intron 11 variant who manifests a milder phenotype consistent with Bethlem muscular dystrophy. Characterisation of this individual provides a robust validation for the development of our pseudoexon skipping therapy. We have previously shown that splice-modulating antisense oligomers applied in vitro effectively decreased the abundance of the mutant pseudoexon-containing COL6A1 transcripts to levels comparable to the in vivo scenario of the somatic mosaicism shown here, indicating that this therapeutic approach carries significant translational promise for ameliorating the severe form of UCMD caused by this common recurrent COL6A1 causative variant to a Bethlem muscular dystrophy phenotype.
Collapse
Affiliation(s)
- A. Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Fady Guirguis
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Rotem Orbach
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
- Dana-Dwek Children’s Hospital, Tel Aviv 64239, Israel
| | - Riley M. McCarty
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Apurva Sarathy
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Gina Norato
- Clinical Trials Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | | | - Monkol Lek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London WC1N 1EH, UK
| | - Russell J. Butterfield
- Departments of Neurology and Pediatrics, University of Utah, Salt Lake City, UT 84132, USA
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg 79110, Germany
| | - Andrés Nascimento
- Neuromuscular Unit, Neuropediatrics Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu. CIBERER ISCIII. Barcelona 08950, Spain
| | - Daniel Natera-de Benito
- Neuromuscular Unit, Neuropediatrics Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu. CIBERER ISCIII. Barcelona 08950, Spain
| | - Susana Quijano-Roy
- Garches Neuromuscular Reference Center, Child Neurology and ICU Department, APHP Raymond Poincare University Hospital (UVSQ Paris Saclay), Garches 92380, France
| | - Tanya Stojkovic
- Centre de Référence des Maladies Neuromusculaires Nord/Est/Île-de-France, Institut de Myologie, Hôpital Pitié-Salpêtrière, AP-HP, Paris 75013, France
| | - Luciano Merlini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40126, Italy
| | - Giacomo Comi
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Monique Ryan
- Department of Neurology, The Royal Children’s Hospital, Parkville, VIC 3052, Australia
| | - Denise McDonald
- Department of Neurodisability, Children’s Health Ireland at Tallaght, Dublin 24 Ireland
| | - Pinki Munot
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London WC1N 1EH, UK
| | - Grace Yoon
- Department of Pediatrics, Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Edward Leung
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| | - Erika Finanger
- Department of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Meganne E. Leach
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
- Department of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - James Collins
- Divisions of Neurology and Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Cuixia Tian
- Divisions of Neurology and Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Payam Mohassel
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Sarah B. Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Dimah Saade
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Benjamin T. Cocanougher
- Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, NC 27710, USA
| | - Mary-Lynn Chu
- Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Mena Scavina
- Division of Neurology, Nemours Children’s Hospital Delaware, Wilmington, DE 19803, USA
| | - Carla Grosmann
- Department of Neurology, Rady Children’s Hospital University of California San Diego, San Diego, CA 92123, USA
| | - Randal Richardson
- Department of Neurology, Gillette Children’s Specialty Healthcare, St Paul, MN 55101, USA
| | - Brian D. Kossak
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, NH 03766, USA
| | - Sidney M. Gospe
- Department of Neurology and Pediatrics, University of Washington, Seattle, WA 98105, USA
| | - Vikram Bhise
- Departments of Pediatrics and Neurology, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Gita Taurina
- Children’s Clinical University Hospital, Medical Genetics and Prenatal Diagnostic Clinic, Riga 1004, Latvia
| | - Baiba Lace
- Riga East Clinical University, Institute of Clinical and Preventive Medicine of the University of Latvia, Riga 1586, Latvia
| | - Monica Troncoso
- Pediatric Neuropsychiatry Service, Hospital Clínico San Borja Arriarán, Pediatric Department, Universidad de Chile, Santiago 1234, Chile
| | - Mordechai Shohat
- The Genomics Unit, Sheba Cancer Research Center, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Adel Shalata
- The Simon Winter Institute for Human Genetics, Bnai Zion Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Sophelia H.S. Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Manu Jokela
- Clinical Neurosciences, University of Turku, Turku, Finland and Neurocenter, Turku University Hospital, Turku 20520, Finland
- Neuromuscular Research Center, Tampere University and Tampere University Hospital, Tampere 33101, Finland
| | - Johanna Palmio
- Neuromuscular Research Center, Tampere University and Tampere University Hospital, Tampere 33101, Finland
| | - Göknur Haliloğlu
- Division of Pediatric Neurology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara 06230, Turkey
| | - Cristina Jou
- Pathology department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona 08950, Spain
| | - Corine Gartioux
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris 75013, France
| | | | - Carolin D. Freiburg
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Alvise Schiavinato
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Haiyan Zhou
- National Institute of Health Research, Great Ormond Street Hospital Biomedical Research Centre, Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sara Aguti
- Neurodegenerative Disease Department, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Yoram Nevo
- Institute of Pediatric Neurology, Schneider Children’s Medical Center of Israel, Petach Tikva, Israel, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Cecilia Jimenez-Mallebrera
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Institut de Recerca Sant Joan de Déu, Barcelona 08950, Spain
| | - Shireen R. Lamandé
- Department of Paediatrics, University of Melbourne, The Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
| | - Valérie Allamand
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris 75013, France
| | - Francesca Gualandi
- Unit of Medical Genetics, Department of Medical Sciences and Department of Mother and Child, University Hospital S. Anna Ferrara, Ferrara 44121, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences and Department of Mother and Child, University Hospital S. Anna Ferrara, Ferrara 44121, Italy
| | | | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University; Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Raimund Wagener
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Disorders, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London WC1N 1EH, UK
- National Institute of Health Research, Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Gregory CA, Ma J, Lomeli S. The coordinated activities of collagen VI and XII in maintenance of tissue structure, function and repair: evidence for a physical interaction. Front Mol Biosci 2024; 11:1376091. [PMID: 38606288 PMCID: PMC11007232 DOI: 10.3389/fmolb.2024.1376091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/14/2024] [Indexed: 04/13/2024] Open
Abstract
Collagen VI and collagen XII are structurally complex collagens of the extracellular matrix (ECM). Like all collagens, type VI and XII both possess triple-helical components that facilitate participation in the ECM network, but collagen VI and XII are distinct from the more abundant fibrillar collagens in that they also possess arrays of structurally globular modules with the capacity to propagate signaling to attached cells. Cell attachment to collagen VI and XII is known to regulate protective, proliferative or developmental processes through a variety of mechanisms, but a growing body of genetic and biochemical evidence suggests that at least some of these phenomena may be potentiated through mechanisms that require coordinated interaction between the two collagens. For example, genetic studies in humans have identified forms of myopathic Ehlers-Danlos syndrome with overlapping phenotypes that result from mutations in either collagen VI or XII, and biochemical and cell-based studies have identified accessory molecules that could form bridging interactions between the two collagens. However, the demonstration of a direct or ternary structural interaction between collagen VI or XII has not yet been reported. This Hypothesis and Theory review article examines the evidence that supports the existence of a functional complex between type VI and XII collagen in the ECM and discusses potential biological implications.
Collapse
Affiliation(s)
- Carl A. Gregory
- Department of Medical Physiology, Texas A&M School of Medicine, Bryan, TX, United States
| | | | | |
Collapse
|
24
|
Bolduc V, Guirguis F, Lubben B, Trank L, Silverstein S, Brull A, Nalls M, Cheng J, Garrett L, Bönnemann CG. A humanized knock-in Col6a1 mouse recapitulates a deep-intronic splice-activating variant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.581572. [PMID: 38585878 PMCID: PMC10996637 DOI: 10.1101/2024.03.21.581572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Antisense therapeutics such as splice-modulating antisense oligonucleotides (ASOs) are promising tools to treat diseases caused by splice-altering intronic variants. However, their testing in animal models is hampered by the generally poor sequence conservation of the intervening sequences between human and other species. Here we aimed to model in the mouse a recurrent, deep-intronic, splice-activating, COL6A1 variant, associated with a severe form of Collagen VI-related muscular dystrophies (COL6-RDs), for the purpose of testing human-ready antisense therapeutics in vivo. The variant, c.930+189C>T, creates a donor splice site and inserts a 72-nt-long pseudoexon, which, when translated, acts in a dominant-negative manner, but which can be skipped with ASOs. We created a unique humanized mouse allele (designated as "h"), in which a 1.9 kb of the mouse genomic region encoding the amino-terminus (N-) of the triple helical (TH) domain of collagen a1(VI) was swapped for the human orthologous sequence. In addition, we also created an allele that carries the c.930+189C>T variant on the same humanized knock-in sequence (designated as "h+189T"). We show that in both models, the human exons are spliced seamlessly with the mouse exons to generate a chimeric mouse-human collagen a1(VI) protein. In homozygous Col6a1 h+189T/h+189T mice, the pseudoexon is expressed at levels comparable to those observed in heterozygous patients' muscle biopsies. While Col6a1h/h mice do not show any phenotype compared to wildtype animals, Col6a1 h/h+189T and Col6a1 h+189T/h+189T mice have smaller muscle masses and display grip strength deficits detectable as early as 4 weeks of age. The pathogenic h+189T humanized knock-in mouse allele thus recapitulates the pathogenic splicing defects seen in patients' biopsies and allows testing of human-ready precision antisense therapeutics aimed at skipping the pseudoexon. Given that the COL6A1 N-TH region is a hot-spot for COL6-RD variants, the humanized knock-in mouse model can be utilized as a template to introduce other COL6A1 pathogenic variants. This unique humanized mouse model thus represents a valuable tool for the development of antisense therapeutics for COL6-RDs.
Collapse
Affiliation(s)
- Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Fady Guirguis
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Berit Lubben
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Lindsey Trank
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Sarah Silverstein
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Astrid Brull
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Matthew Nalls
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Jun Cheng
- NHGRI Transgenic and Gene Editing Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Lisa Garrett
- NHGRI Transgenic and Gene Editing Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| |
Collapse
|
25
|
Bolduc V, Sizov K, Brull A, Esposito E, Chen GS, Uapinyoying P, Sarathy A, Johnson K, Bönnemann CG. Allele-specific CRISPR/Cas9 editing inactivates a single nucleotide variant associated with collagen VI muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586265. [PMID: 38585815 PMCID: PMC10996683 DOI: 10.1101/2024.03.22.586265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The application of allele-specific gene editing tools can expand the therapeutic options for dominant genetic conditions, either via gene correction or via allelic gene inactivation in situations where haploinsufficiency is tolerated. Here, we used allele-targeted CRISPR/Cas9 guide RNAs (gRNAs) to introduce inactivating frameshifting indels at a single nucleotide variant in the COL6A1 gene (c.868G>A; G290R), a variant that acts as dominant negative and that is associated with a severe form of congenital muscular dystrophy. We expressed spCas9 along with allele-targeted gRNAs, without providing a repair template, in primary fibroblasts derived from four patients and one control subject. Amplicon deep-sequencing for two gRNAs tested showed that single nucleotide deletions accounted for the majority of indels introduced. While activity of the two gRNAs was greater at the G290R allele, both gRNAs were also active at the wild-type allele. To enhance allele-selectivity, we introduced deliberate additional mismatches to one gRNA. One of these optimized gRNAs showed minimal activity at the WT allele, while generating productive edits and improving collagen VI matrix in cultured patient fibroblasts. This study strengthens the potential of gene editing to treat dominant-negative disorders, but also underscores the challenges in achieving allele selectivity with gRNAs.
Collapse
Affiliation(s)
- Véronique Bolduc
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Katherine Sizov
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Astrid Brull
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eric Esposito
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Grace S Chen
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Prech Uapinyoying
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Center for Genetic Medicine Research, Children's National Research and Innovation Campus, Children's National Hospital, Washington, DC, 20012, USA
| | - Apurva Sarathy
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kory Johnson
- Bioinformatics Core, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Carsten G Bönnemann
- Neurogenetics and Neuromuscular Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Metti S, Da Ros F, Toniato G, Cescon M, Bonaldo P. Native collagen VI delays early muscle stem cell differentiation. J Cell Sci 2024; 137:jcs261419. [PMID: 38224152 PMCID: PMC10911284 DOI: 10.1242/jcs.261419] [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: 06/17/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024] Open
Abstract
Adult muscle stem cells (MuSCs) are critical for muscle homeostasis and regeneration, and their behavior relies on a finely regulated niche made of specific extracellular matrix (ECM) components and soluble factors. Among ECM proteins, collagen VI (Col6) influences the mechanical properties of the niche and, in turn, MuSC self-renewal capabilities. Here, we investigated whether Col6 can exert a direct function as a biochemical signal for regulating the stemness and differentiation of murine MuSCs and myoblasts. Native Col6, but not its pepsin-resistant fragment, counteracts the early differentiation of myogenic cells by reducing the expression of differentiation marker genes and preserving stemness features, with inhibition of the canonical Wnt pathway. Our data indicate that extracellular Col6 acts as a soluble ligand in delaying early myogenic differentiation by regulating intracellular signals involved in adult myogenesis.
Collapse
Affiliation(s)
- Samuele Metti
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Francesco Da Ros
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Giorgia Toniato
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| |
Collapse
|
27
|
Morel V, Audic F, Tardy C, Verschueren A, Attarian S, Nguyen K, Salort-Campana E, Krahn M, Chabrol B, Gorokhova S. Retrospective clinical and genetic analysis of COL6-RD patients with a long-term follow-up at a single French center. Front Genet 2023; 14:1242277. [PMID: 38155714 PMCID: PMC10753780 DOI: 10.3389/fgene.2023.1242277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Collagen type VI-related dystrophies (COL6-RD) are rare diseases with a wide phenotypic spectrum ranging from severe Ullrich's congenital muscular dystrophy Ullrich congenital muscular dystrophy to much milder Bethlem myopathy Both dominant and recessive forms of COL6-RD are caused by pathogenic variants in three collagen VI genes (COL6A1, COL6A2 and COL6A3). The prognosis of these diseases is variable and difficult to predict during early disease stages, especially since the genotype-phenotype correlation is not always clear. For this reason, studies with long-term follow-up of patients with genetically confirmed COL6-RD are still needed. In this study, we present phenotypic and genetic data from 25 patients (22 families) diagnosed with COL6-RD and followed at a single French center, in both adult and pediatric neurology departments. We describe three novel pathogenic variants and identify COL6A2:c.1970-9G>A as the most frequent variant in our series (29%). We also observe an accelerated progression of the disease in a subgroup of patients. This large series of rare disease patients provides essential information on phenotypic variability of COL6-RD patients as well as on frequency of pathogenic COL6A gene variants in Southern France, thus contributing to the phenotypic and genetic description of Collagen type VI-related dystrophies.
Collapse
Affiliation(s)
- Victor Morel
- Département de Génétique Médicale, Hôpital de la Timone, Marseille, Provence-Alpes-Côte d’Azur, France
| | - Frédérique Audic
- Service de Neuropédiatrie, Centre de Référence des Maladies Neuromusculaires de l’enfant PACARARE, CHU Timone, APHM, Marseille, France
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix Marseille University, Marseille, France
| | - Charlotte Tardy
- Département de Génétique Médicale, Hôpital de la Timone, Marseille, Provence-Alpes-Côte d’Azur, France
| | - Annie Verschueren
- Centre de Référence des Maladies Neuromusculaires et de la SLA, ERN-NMD, CHU Timone, APHM, Marseille, France
| | - Shahram Attarian
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix Marseille University, Marseille, France
- Centre de Référence des Maladies Neuromusculaires et de la SLA, ERN-NMD, CHU Timone, APHM, Marseille, France
| | - Karine Nguyen
- Département de Génétique Médicale, Hôpital de la Timone, Marseille, Provence-Alpes-Côte d’Azur, France
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix Marseille University, Marseille, France
| | - Emmanuelle Salort-Campana
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix Marseille University, Marseille, France
- Centre de Référence des Maladies Neuromusculaires et de la SLA, ERN-NMD, CHU Timone, APHM, Marseille, France
| | - Martin Krahn
- Département de Génétique Médicale, Hôpital de la Timone, Marseille, Provence-Alpes-Côte d’Azur, France
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix Marseille University, Marseille, France
| | - Brigitte Chabrol
- Service de Neuropédiatrie, Centre de Référence des Maladies Neuromusculaires de l’enfant PACARARE, CHU Timone, APHM, Marseille, France
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix Marseille University, Marseille, France
| | - Svetlana Gorokhova
- Département de Génétique Médicale, Hôpital de la Timone, Marseille, Provence-Alpes-Côte d’Azur, France
- Inserm, U1251-MMG, Marseille Medical Genetics, Aix Marseille University, Marseille, France
| |
Collapse
|
28
|
Pascoe JE, Zygmunt A, Ehsan Z, Gurbani N. Sleep in pediatric neuromuscular disorders. Semin Pediatr Neurol 2023; 48:101092. [PMID: 38065635 DOI: 10.1016/j.spen.2023.101092] [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: 08/14/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 12/18/2023]
Abstract
Sleep disordered breathing (SDB) is prevalent among children with neuromuscular disorders (NMD). The combination of respiratory muscle weakness, altered drive, and chest wall distortion due to scoliosis make sleep a stressful state in this population. Symptomatology can range from absent to snoring, nocturnal awakenings, morning headaches, and excessive daytime sleepiness. Sequelae of untreated SDB includes cardiovascular effects, metabolic derangements, and neurocognitive concerns which can be compounded by those innate to the NMD. The clinician should have a low threshold for obtaining polysomnography and recognize the nuances of individual disorders due to disproportionately impacted muscle groups such as hypoventilation in ambulating patients from diaphragm weakness. Non-invasive or invasive ventilation are the mainstay of treatment. In this review we explore the diagnosis and treatment of SDB in children with various NMD.
Collapse
Affiliation(s)
- John E Pascoe
- Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| | - Alexander Zygmunt
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Zarmina Ehsan
- Division of Pulmonary and Sleep Medicine, Children's Mercy-Kansas City, Kansas City, MO, United States; Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Neepa Gurbani
- Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
29
|
Mavrommatis L, Jeong HW, Kindler U, Gomez-Giro G, Kienitz MC, Stehling M, Psathaki OE, Zeuschner D, Bixel MG, Han D, Morosan-Puopolo G, Gerovska D, Yang JH, Kim JB, Arauzo-Bravo MJ, Schwamborn JC, Hahn SA, Adams RH, Schöler HR, Vorgerd M, Brand-Saberi B, Zaehres H. Human skeletal muscle organoids model fetal myogenesis and sustain uncommitted PAX7 myogenic progenitors. eLife 2023; 12:RP87081. [PMID: 37963071 PMCID: PMC10645425 DOI: 10.7554/elife.87081] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
In vitro culture systems that structurally model human myogenesis and promote PAX7+ myogenic progenitor maturation have not been established. Here we report that human skeletal muscle organoids can be differentiated from induced pluripotent stem cell lines to contain paraxial mesoderm and neuromesodermal progenitors and develop into organized structures reassembling neural plate border and dermomyotome. Culture conditions instigate neural lineage arrest and promote fetal hypaxial myogenesis toward limb axial anatomical identity, with generation of sustainable uncommitted PAX7 myogenic progenitors and fibroadipogenic (PDGFRa+) progenitor populations equivalent to those from the second trimester of human gestation. Single-cell comparison to human fetal and adult myogenic progenitor /satellite cells reveals distinct molecular signatures for non-dividing myogenic progenitors in activated (CD44High/CD98+/MYOD1+) and dormant (PAX7High/FBN1High/SPRY1High) states. Our approach provides a robust 3D in vitro developmental system for investigating muscle tissue morphogenesis and homeostasis.
Collapse
Affiliation(s)
- Lampros Mavrommatis
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular EmbryologyBochumGermany
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental BiologyMünsterGermany
- Department of Neurology with Heimer Institute for Muscle Research, University Hospital BergmannsheilBochumGermany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Sequencing Core FacilityMünsterGermany
| | - Urs Kindler
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular EmbryologyBochumGermany
| | - Gemma Gomez-Giro
- Luxembourg Centre for Systems Biomedicine, LCSB, Developmental and Cellular Biology, University of LuxembourgBelvauxLuxembourg
| | - Marie-Cecile Kienitz
- Ruhr University Bochum, Medical Faculty, Department of Cellular PhysiologyBochumGermany
| | - Martin Stehling
- Max Planck Institute for Molecular Biomedicine, Flow Cytometry UnitMünsterGermany
| | - Olympia E Psathaki
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental BiologyMünsterGermany
- Center for Cellular Nanoanalytics Osnabrück, CellNanOs, University of OsnabrückOsnabrückGermany
| | - Dagmar Zeuschner
- Max Planck Institute for Molecular Biomedicine, Electron Microscopy UnitMünsterGermany
| | - M Gabriele Bixel
- Max Planck Institute for Molecular Biomedicine, Department of Tissue MorphogenesisMünsterGermany
| | - Dong Han
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental BiologyMünsterGermany
| | - Gabriela Morosan-Puopolo
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular EmbryologyBochumGermany
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biodonostia Health Research InstituteSan SebastiánSpain
| | - Ji Hun Yang
- School of Mechanical Engineering, Korea UniversitySeoulRepublic of Korea
- R&D Research Center, Next & Bio IncSeoulRepublic of Korea
| | - Jeong Beom Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST)UlsanRepublic of Korea
| | - Marcos J Arauzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research InstituteSan SebastiánSpain
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine, LCSB, Developmental and Cellular Biology, University of LuxembourgBelvauxLuxembourg
| | - Stephan A Hahn
- Ruhr University Bochum, Medical Faculty, Department of Molecular GI OncologyBochumGermany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue MorphogenesisMünsterGermany
- Westphalian Wilhelms University Münster, Medical FacultyMünsterGermany
| | - Hans R Schöler
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental BiologyMünsterGermany
| | - Matthias Vorgerd
- Department of Neurology with Heimer Institute for Muscle Research, University Hospital BergmannsheilBochumGermany
| | - Beate Brand-Saberi
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular EmbryologyBochumGermany
| | - Holm Zaehres
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular EmbryologyBochumGermany
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental BiologyMünsterGermany
| |
Collapse
|
30
|
Jankelunas L, Murthy VD, Chen AV, Minor KM, Friedenberg SG, Cullen JN, Guo LT, Mickelson JR, Shelton GD. Novel COL6A3 frameshift variant in American Staffordshire Terrier dogs with Ullrich-like congenital muscular dystrophy. J Vet Intern Med 2023; 37:2504-2509. [PMID: 37706358 PMCID: PMC10658572 DOI: 10.1111/jvim.16862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
Two (male and female) 10-month-old American Staffordshire Terrier littermates presented for progressive weakness, joint contracture, and distal limb joint hyperlaxity beginning around 6 months of age. Neurological examination, serum creatine kinase activity, infectious disease titers, cerebrospinal fluid analysis, and electrodiagnostic testing were performed. Muscle biopsies were collected for histopathology and immunofluorescence staining for localization of dystrophy associated proteins. Whole-genome sequencing (WGS) was performed on 1 affected dog. Variants were compared to a database of 671 unaffected dogs of multiple breeds. Histopathology confirmed a dystrophic phenotype and immunofluorescence staining of muscle cryosections revealed an absence of staining for collagen-6. WGS identified a homozygous 1 bp deletion in the COL6A3 gene, unique to the first affected dog. Sanger sequencing confirmed the homozygous presence of the frameshift variant in both affected dogs. This report describes the clinical features and most likely genetic basis of an Ullrich-like recessively inherited form of congenital muscular dystrophy in American Staffordshire Terriers.
Collapse
Affiliation(s)
- Leanne Jankelunas
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| | - Vishal D. Murthy
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| | - Annie V. Chen
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineWashington State UniversityPullmanWashingtonUSA
| | - Katie M. Minor
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - Steven G. Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - Jonah N. Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - Ling T. Guo
- Department of Pathology, School of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - James R. Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary MedicineUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - G. Diane Shelton
- Department of Pathology, School of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
31
|
Su W, Liu Y, Lam A, Hao X, Baudry M, Bi X. Contextual fear memory impairment in Angelman syndrome model mice is associated with altered transcriptional responses. Sci Rep 2023; 13:18647. [PMID: 37903805 PMCID: PMC10616231 DOI: 10.1038/s41598-023-45769-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023] Open
Abstract
Angelman syndrome (AS) is a rare neurogenetic disorder caused by UBE3A deficiency and characterized by severe developmental delay, cognitive impairment, and motor dysfunction. In the present study, we performed RNA-seq on hippocampal samples from both wildtype (WT) and AS male mice, with or without contextual fear memory recall. There were 281 recall-associated differentially expressed genes (DEGs) in WT mice and 268 DEGs in AS mice, with 129 shared by the two genotypes. Gene ontology analysis showed that extracellular matrix and stimulation-induced response genes were prominently enriched in recall-associated DEGs in WT mice, while nuclear acid metabolism and tissue development genes were highly enriched in those from AS mice. Further analyses showed that the 129 shared DEGs belonged to nuclear acid metabolism and tissue development genes. Unique recall DEGs in WT mice were enriched in biological processes critical for synaptic plasticity and learning and memory, including the extracellular matrix network clustered around fibronectin 1 and collagens. In contrast, AS-specific DEGs were not enriched in any known pathways. These results suggest that memory recall in AS mice, while altering the transcriptome, fails to recruit memory-associated transcriptional programs, which could be responsible for the memory impairment in AS mice.
Collapse
Affiliation(s)
- Wenyue Su
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Yan Liu
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Aileen Lam
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA
| | - Xiaoning Hao
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA
| | - Michel Baudry
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA.
| |
Collapse
|
32
|
Zeng L, Tang Y, Zhang Y, Yue L, Ma G, Ye X, Yang L, Chen K, Zhou Q. The molecular mechanism underlying dermatomyositis related interstitial lung disease: evidence from bioinformatic analysis and in vivo validation. Front Immunol 2023; 14:1288098. [PMID: 37928522 PMCID: PMC10622801 DOI: 10.3389/fimmu.2023.1288098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Background Dermatomyositis (DM) is an autoimmune and inflammatory disease that can affect the lungs, causing interstitial lung diseases (ILD). However, the exact pathophysiological mechanisms underlying DM-ILD are unknown. Idiopathic pulmonary fibrosis (IPF) belongs to the broader spectrum of ILD and evidence shows that common pathologic pathways might lie between IPF and DM-ILD. Methods We retrieved gene expression profiles of DM and IPF from the Gene Expression Omnibus (GEO) and utilized weighted gene co-expression network analysis (WGCNA) to reveal their co-expression modules. We then performed a differentially expressed gene (DEG) analysis to identify common DEGs. Enrichment analyses were employed to uncover the hidden biological pathways. Additionally, we conducted protein-protein interaction (PPI) networks analysis, cluster analysis, and successfully found the hub genes, whose levels were further validated in DM-ILD patients. We also examined the relationship between hub genes and immune cell abundance in DM and IPF. Finally, we conducted a common transcription factors (TFs)-genes network by NetworkAnalyst. Results WGCNA revealed 258 intersecting genes, while DEG analysis identified 66 shared genes in DM and IPF. All of these genes were closely related to extracellular matrix and structure, cell-substrate adhesion, and collagen metabolism. Four hub genes (POSTN, THBS2, COL6A1, and LOXL1) were derived through intersecting the top 30 genes of the WGCNA and DEG sets. They were validated as active transcripts and showed diagnostic values for DM and IPF. However, ssGSEA revealed distinct infiltration patterns in DM and IPF. These four genes all showed a positive correlation with immune cells abundance in DM, but not in IPF. Finally, we identified one possible key transcription factor, MYC, that interact with all four hub genes. Conclusion Through bioinformatics analysis, we identified common hub genes and shared molecular pathways underlying DM and IPF, which provides valuable insights into the intricate mechanisms of these diseases and offers potential targets for diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Li Zeng
- Department of Neurology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yiping Tang
- Department of Internal Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yichen Zhang
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Yue
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gang Ma
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xumin Ye
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Southwest Medical University, Luzhou, China
| | - Lijing Yang
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Kai Chen
- Department of Neurology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
33
|
Freiburg CD, Solomon-Degefa H, Freiburg P, Mörgelin M, Bolduc V, Schmitz S, Ala P, Muntoni F, Behrmann E, Bönnemann CG, Schiavinato A, Paulsson M, Wagener R. The UCMD-Causing COL6A1 ( c.930 + 189 C > T) Intron Mutation Leads to the Secretion and Aggregation of Single Mutated Collagen VI α1 Chains. Hum Mutat 2023; 2023:6892763. [PMID: 40225172 PMCID: PMC11919215 DOI: 10.1155/2023/6892763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 04/05/2025]
Abstract
Collagen VI is a unique member of the collagen family. Its assembly is a complex multistep process and the vulnerability of the process is manifested in muscular diseases. Mutations in COL6A1, COL6A2, and COL6A3 lead to the severe Ullrich Congenital Muscular Dystrophy (UCMD) and a spectrum of disease of varying severity including the milder Bethlem muscular dystrophy. The recently identified dominant intronic mutation in COL6A1 (c.930 + 189C > T) leads to the partial in-frame insertion of a pseudoexon between exon 11 and exon 12. The pseudoexon is translated into 24 amino acid residues in the N-terminal region of the triple helix and results in the interruption of the typical G-X-Y motif. This recurrent de novo mutation leads to UCMD with a severe progression within the first decade of life. Here, we demonstrate that a mutation-specific antibody detects the mutant chain colocalizing with wild type collagen VI in the endomysium in patient muscle. Surprisingly, in the cell culture of patient dermal fibroblasts, the mutant chain is secreted as a single α chain, while in parallel, normal collagen VI tetramers are assembled with the wild-type α1 chain. The mutant chain cannot be incorporated into collagen VI tetramers but forms large aggregates in the extracellular matrix that may retain the ability to interact with collagen VI and potentially with other molecules. Also, α1 chain-deficient WI-26 VA4 cells transfected with the mutant α1 chain do not assemble collagen VI tetramers but, instead, form aggregates. Interestingly, both the wild type and the mutant single α1 chains form amorphous aggregates when expressed in HEK293 cells in the absence of α2 and α3 chains. The detection of aggregated, assembly incompetent, mutant collagen VI α1 chains provides novel insights into the disease pathophysiology of UCMD patients with the COL6A1 (c.930 + 189C > T) mutation.
Collapse
Affiliation(s)
- Carolin D. Freiburg
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | | | - Patrick Freiburg
- Institute of Biochemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | | | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Sebastian Schmitz
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Pierpaolo Ala
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital NIHR Biomedical Research Centre, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital NIHR Biomedical Research Centre, London, UK
| | - Elmar Behrmann
- Institute of Biochemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Alvise Schiavinato
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), Cologne, Germany
| | - Mats Paulsson
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), Cologne, Germany
- Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| | - Raimund Wagener
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
34
|
Mohassel P, Rooney J, Zou Y, Johnson K, Norato G, Hearn H, Nalls MA, Yun P, Ogata T, Silverstein S, Sleboda DA, Roberts TJ, Rifkin DB, Bönnemann CG. Collagen type VI regulates TGFβ bioavailability in skeletal muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.22.545964. [PMID: 38586035 PMCID: PMC10996771 DOI: 10.1101/2023.06.22.545964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Collagen VI-related disorders (COL6-RDs) are a group of rare muscular dystrophies caused by pathogenic variants in collagen VI genes (COL6A1, COL6A2, and COL6A3). Collagen type VI is a heterotrimeric, microfibrillar component of the muscle extracellular matrix (ECM), predominantly secreted by resident fibroadipogenic precursor cells in skeletal muscle. The absence or mislocalizatoion of collagen VI in the ECM underlies the non-cell autonomous dysfunction and dystrophic changes in skeletal muscle with an as of yet elusive direct mechanistic link between the ECM and myofiber dysfunction. Here, we conduct a comprehensive natural history and outcome study in a novel mouse model of COL6-RDs (Col6a2 -/- mice) using standardized (Treat-NMD) functional, histological, and physiologic parameter. Notably, we identify a conspicuous dysregulation of the TGFβ pathway early in the disease process and propose that the collagen VI deficient matrix is not capable of regulating the dynamic TGFβ bioavailability at baseline and also in response to muscle injury. Thus, we propose a new mechanism for pathogenesis of the disease that links the ECM regulation of TGFβ with downstream skeletal muscle abnormalities, paving the way for developing and validating therapeutics that target this pathway.
Collapse
Affiliation(s)
- Payam Mohassel
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jachinta Rooney
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
| | - Yaqun Zou
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
| | - Kory Johnson
- Bioinformatics Section, Intramural Information Technology & Bioinformatics Program, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Gina Norato
- Clinical Trials Unit, National Institutes of Health, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Hailey Hearn
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew A Nalls
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
| | - Pomi Yun
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
| | - Tracy Ogata
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
| | - Sarah Silverstein
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
| | - David A Sleboda
- Department of Ecology and Evolutionary Biology, University of California, Irvine, Irvine, CA, USA
| | - Thomas J Roberts
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Daniel B Rifkin
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Carsten G Bönnemann
- National Institutes of Health, National Institute of Neurological Disorders and Stroke, Neuromuscular and Neurogenetic Disorders of Childhood Section, Bethesda, MD, USA
| |
Collapse
|
35
|
Starace M, Pampaloni F, Bruni F, Quadrelli F, Cedirian S, Baraldi C, Misciali C, Di Martino A, Sabatelli P, Merlini L, Piraccini BM. Alopecia in Patients with Collagen VI-Related Myopathies: A Novel/Unrecognized Scalp Phenotype. Int J Mol Sci 2023; 24:ijms24076678. [PMID: 37047652 PMCID: PMC10095448 DOI: 10.3390/ijms24076678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/01/2023] [Accepted: 04/01/2023] [Indexed: 04/14/2023] Open
Abstract
Collagen VI-related myopathies are characterized by severe muscle involvement and skin involvement (keratosis pilaris and impaired healing with the development of abnormal scars, especially keloids). Scalp involvement and hair loss have not been reported among cutaneous changes associated with collagen VI mutations. The aim of this study is to describe the clinical, trichoscopic, and histological findings of the scalp changes in patients affected by COL VI mutations and to estimate their prevalence. Patients with Ullrich congenital muscular dystrophy were enrolled and underwent clinical and trichoscopic examinations and a scalp biopsy for histopathology. Five patients were enrolled, and all complained of hair loss and scalp itching. One patient showed yellow interfollicular scales with erythema and dilated, branched vessels, and the histological findings were suggestive of scalp psoriasis. Two patients presented with scarring alopecia patches on the vertex area, and they were histologically diagnosed with folliculitis decalvans. The last two patients presented with scaling and hair thinning, but they were both diagnosed with folliculitis and perifolliculitis. Ten more patients answered to a "scalp involvement questionnaire", and six of them confirmed to have or have had scalp disorders and/or itching. Scalp involvement can be associated with COL VI mutations and should be investigated.
Collapse
Affiliation(s)
- Michela Starace
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Francesca Pampaloni
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Francesca Bruni
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Federico Quadrelli
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Stephano Cedirian
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Carlotta Baraldi
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Cosimo Misciali
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Alberto Di Martino
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
- Clinica Ortopedica e Traumatologica I, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Patrizia Sabatelli
- Unit of Bologna, CNR-Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Luciano Merlini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Bianca Maria Piraccini
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
36
|
Zanotti S, Magri F, Salani S, Napoli L, Ripolone M, Ronchi D, Fortunato F, Ciscato P, Velardo D, D’Angelo MG, Gualandi F, Nigro V, Sciacco M, Corti S, Comi GP, Piga D. Extracellular Matrix Disorganization and Sarcolemmal Alterations in COL6-Related Myopathy Patients with New Variants of COL6 Genes. Int J Mol Sci 2023; 24:5551. [PMID: 36982625 PMCID: PMC10059973 DOI: 10.3390/ijms24065551] [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: 11/30/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Collagen VI is a heterotrimeric protein expressed in several tissues and involved in the maintenance of cell integrity. It localizes at the cell surface, creating a microfilamentous network that links the cytoskeleton to the extracellular matrix. The heterotrimer consists of three chains encoded by COL6A1, COL6A2 and COL6A3 genes. Recessive and dominant molecular defects cause two main disorders, the severe Ullrich congenital muscular dystrophy and the relatively mild and slowly progressive Bethlem myopathy. We analyzed the clinical aspects, pathological features and mutational spectrum of 15 COL6-mutated patients belonging to our cohort of muscular dystrophy probands. Patients presented a heterogeneous phenotype ranging from severe forms to mild adult-onset presentations. Molecular analysis by NGS detected 14 different pathogenic variants, three of them so far unreported. Two changes, localized in the triple-helical domain of COL6A1, were associated with a more severe phenotype. Histological, immunological and ultrastructural techniques were employed for the validation of the genetic variants; they documented the high variability in COL6 distribution and the extracellular matrix disorganization, highlighting the clinical heterogeneity of our cohort. The combined use of these different technologies is pivotal in the diagnosis of COL6 patients.
Collapse
Affiliation(s)
- Simona Zanotti
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Magri
- Neurology Unit, Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sabrina Salani
- Neurology Unit, Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Laura Napoli
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Michela Ripolone
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Dario Ronchi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy
| | - Francesco Fortunato
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy
| | - Patrizia Ciscato
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Daniele Velardo
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | | | - Francesca Gualandi
- Medical Genetics Unit, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy
| | - Vincenzo Nigro
- Dipartimento di Medicina di Precisione, “Luigi Vanvitelli” University of Campania and Telethon Institute of Genetics and Medicine (TIGEM), 81100 Naples, Italy
| | - Monica Sciacco
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Neurology Unit, Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefania Corti
- Neurology Unit, Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy
| | - Giacomo Pietro Comi
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy
| | - Daniela Piga
- Neurology Unit, Department of Neuroscience Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
37
|
Di Martino A, Cescon M, D’Agostino C, Schilardi F, Sabatelli P, Merlini L, Faldini C. Collagen VI in the Musculoskeletal System. Int J Mol Sci 2023; 24:5095. [PMID: 36982167 PMCID: PMC10049728 DOI: 10.3390/ijms24065095] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/10/2023] Open
Abstract
Collagen VI exerts several functions in the tissues in which it is expressed, including mechanical roles, cytoprotective functions with the inhibition of apoptosis and oxidative damage, and the promotion of tumor growth and progression by the regulation of cell differentiation and autophagic mechanisms. Mutations in the genes encoding collagen VI main chains, COL6A1, COL6A2 and COL6A3, are responsible for a spectrum of congenital muscular disorders, namely Ullrich congenital muscular dystrophy (UCMD), Bethlem myopathy (BM) and myosclerosis myopathy (MM), which show a variable combination of muscle wasting and weakness, joint contractures, distal laxity, and respiratory compromise. No effective therapeutic strategy is available so far for these diseases; moreover, the effects of collagen VI mutations on other tissues is poorly investigated. The aim of this review is to outline the role of collagen VI in the musculoskeletal system and to give an update about the tissue-specific functions revealed by studies on animal models and from patients' derived samples in order to fill the knowledge gap between scientists and the clinicians who daily manage patients affected by collagen VI-related myopathies.
Collapse
Affiliation(s)
- Alberto Di Martino
- I Orthopedic and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Claudio D’Agostino
- I Orthopedic and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy
| | - Francesco Schilardi
- I Orthopedic and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy
| | - Patrizia Sabatelli
- Unit of Bologna, CNR-Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Luciano Merlini
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy
| | - Cesare Faldini
- I Orthopedic and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy
| |
Collapse
|
38
|
Lee SA, Hong JM, Lee JH, Choi YC, Park HJ. Transcriptome profiling of skeletal muscles from Korean patients with Bethlem myopathy. Medicine (Baltimore) 2023; 102:e33122. [PMID: 36862922 PMCID: PMC9981387 DOI: 10.1097/md.0000000000033122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Bethlem myopathy is one of the collagens VI-related muscular dystrophies caused by mutations in the collagen VI genes. The study was designed to analyze the gene expression profiles in the skeletal muscle of patients with Bethlem myopathy. Six skeletal muscle samples from 3 patients with Bethlem myopathy and 3 control subjects were analyzed by RNA-sequencing. 187 transcripts were significantly differentially expressed, with 157 upregulated and 30 downregulated transcripts in the Bethlem group. Particularly, 1 (microRNA-133b) was considerably upregulated, and 4 long intergenic non-protein coding RNAs, LINC01854, MBNL1-AS1, LINC02609, and LOC728975, were significantly downregulated. We categorized differentially expressed gene using Gene Ontology and showed that Bethlem myopathy is strongly associated with the organization of extracellular matrix (ECM). Kyoto Encyclopedia of Genes and Genomes pathway enrichment reflected themes with significant enrichment of the ECM-receptor interaction (hsa04512), complement and coagulation cascades (hsa04610), and focal adhesion (hsa04510). We confirmed that Bethlem myopathy is strongly associated with the organization of ECM and the wound healing process. Our results demonstrate transcriptome profiling of Bethlem myopathy, and provide new insights into the path mechanism of Bethlem myopathy associated with non-protein coding RNAs.
Collapse
Affiliation(s)
- Seung-Ah Lee
- Department of Neurology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Yangcheon-gu, Seoul, Republic of Korea
| | - Ji-Man Hong
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi-do, Republic of Korea
| | - Jung Hwan Lee
- Department of Neurology, Seoul St. Mary’s Hospital, College of Medicine, Seocho-gu, Seoul, Republic of Korea
| | - Young-Chul Choi
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyung Jun Park
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- * Correspondence: Hyung Jun Park, Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, Korea (e-mail: )
| |
Collapse
|
39
|
Abbonante V, Malara A, Chrisam M, Metti S, Soprano P, Semplicini C, Bello L, Bozzi V, Battiston M, Pecci A, Pegoraro E, De Marco L, Braghetta P, Bonaldo P, Balduini A. Lack of COL6/collagen VI causes megakaryocyte dysfunction by impairing autophagy and inducing apoptosis. Autophagy 2023; 19:984-999. [PMID: 35857791 PMCID: PMC9980446 DOI: 10.1080/15548627.2022.2100105] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Endoplasmic reticulum stress is an emerging significant player in the molecular pathology of connective tissue disorders. In response to endoplasmic reticulum stress, cells can upregulate macroautophagy/autophagy, a fundamental cellular homeostatic process used by cells to degrade and recycle proteins or remove damaged organelles. In these scenarios, autophagy activation can support cell survival. Here we demonstrated by in vitro and in vivo approaches that megakaryocytes derived from col6a1-⁄- (collagen, type VI, alpha 1) null mice display increased intracellular retention of COL6 polypeptides, endoplasmic reticulum stress and apoptosis. The unfolded protein response is activated in col6a1-⁄- megakaryocytes, as evidenced by the upregulation of molecular chaperones, by the increased splicing of Xbp1 mRNA and by the higher level of the pro-apoptotic regulator DDIT3/CHOP. Despite the endoplasmic reticulum stress, basal autophagy is impaired in col6a1-⁄- megakaryocytes, which show lower BECN1 levels and reduced autophagosome maturation. Starvation and rapamycin treatment rescue the autophagic flux in col6a1-⁄- megakaryocytes, leading to a decrease in intracellular COL6 polypeptide retention, endoplasmic reticulum stress and apoptosis. Furthermore, megakaryocytes cultured from peripheral blood hematopoietic progenitors of patients affected by Bethlem myopathy and Ullrich congenital muscular dystrophy, two COL6-related disorders, displayed increased apoptosis, endoplasmic reticulum stress and impaired autophagy. These data demonstrate that genetic disorders of collagens, endoplasmic reticulum stress and autophagy regulation in megakaryocytes may be interrelated.Abbreviations: 7-AAD: 7-amino-actinomycin D; ATF: activating transcriptional factor; BAX: BCL2 associated X protein; BCL2: B cell leukemia/lymphoma 2; BCL2L1/Bcl-xL: BCL2-like 1; BM: bone marrow; COL6: collagen, type VI; col6a1-⁄-: mice that are null for Col6a1; DDIT3/CHOP/GADD153: DNA-damage inducible transcript 3; EGFP: enhanced green fluorescent protein; ER: endoplasmic reticulum; reticulophagy: endoplasmic reticulum-selective autophagy; HSPA5/Bip: heat shock protein 5; HSP90B1/GRP94: heat shock protein 90, beta (Grp94), member 1; LAMP2: lysosomal associated membrane protein 2; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; Mk: megakaryocytes; MTOR: mechanistic target of rapamycin kinase; NIMV: noninvasive mechanical ventilation; PI3K: phosphoinositide 3-kinase; PPP1R15A/GADD34: protein phosphatase 1, regulatory subunit 15A; RT-qPCR: reverse transcription-quantitative real-time PCR; ROS: reactive oxygen species; SERPINH1/HSP47: serine (or cysteine) peptidase inhibitor, clade H, member 1; sh-RNA: short hairpin RNA; SOCE: store operated calcium entry; UCMD: Ullrich congenital muscular dystrophy; UPR: unfolded protein response; WIPI2: WD repeat domain, phosphoinositide-interacting 2; WT: wild type; XBP1: X-box binding protein 1.
Collapse
Affiliation(s)
- Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry-Biotechnology and Advanced Diagnostics, IRCCS San Matteo Foundation, Pavia, Italy.,Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry-Biotechnology and Advanced Diagnostics, IRCCS San Matteo Foundation, Pavia, Italy
| | - Martina Chrisam
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Samuele Metti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Paolo Soprano
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry-Biotechnology and Advanced Diagnostics, IRCCS San Matteo Foundation, Pavia, Italy
| | | | - Luca Bello
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Valeria Bozzi
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Monica Battiston
- Department of Translational Research, Stem Cell Unit, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Luigi De Marco
- Department of Translational Research, Stem Cell Unit, CRO Aviano National Cancer Institute, Aviano, Italy.,Department of Molecular and Experimental Medicine, SCRIPPS Research Institute, La Jolla, CA, USA
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Laboratory of Biochemistry-Biotechnology and Advanced Diagnostics, IRCCS San Matteo Foundation, Pavia, Italy.,Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
40
|
Genetic Analysis of HIBM Myopathy-Specific GNE V727M Hotspot Mutation Identifies a Novel COL6A3 Allied Gene Signature That Is Also Deregulated in Multiple Neuromuscular Diseases and Myopathies. Genes (Basel) 2023; 14:genes14030567. [PMID: 36980840 PMCID: PMC10048522 DOI: 10.3390/genes14030567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
The GNE-associated V727M mutation is one of the most prevalent ethnic founder mutations in the Asian HIBM cohort; however, its role in inducing disease phenotype remains largely elusive. In this study, the function of this hotspot mutation was profoundly investigated. For this, V727M mutation-specific altered expression profile and potential networks were explored. The relevant muscular disorder-specific in vivo studies and patient data were further analyzed, and the key altered molecular pathways were identified. Our study found that the GNEV727M mutation resulted in a deregulated lincRNA profile, the majority of which (91%) were associated with a down-regulation trend. Further, in silico analysis of associated targets showed their active role in regulating Wnt, TGF-β, and apoptotic signaling. Interestingly, COL6a3 was found as a key target of these lincRNAs. Further, GSEA analysis showed HIBM patients with variable COL6A3 transcript levels have significant alteration in many critical pathways, including epithelial-mesenchymal-transition, myogenesis, and apoptotic signaling. Interestingly, 12 of the COL6A3 coexpressed genes also showed a similar altered expression profile in HIBM. A similar altered trend in COL6A3 and coexpressed genes were found in in vivo HIBM disease models as well as in multiple other skeletal disorders. Thus, the COL6A3-specific 13 gene signature seems to be altered in multiple muscular disorders. Such deregulation could play a pivotal role in regulating many critical processes such as extracellular matrix organization, cell adhesion, and skeletal muscle development. Thus, investigating this novel COL6A3-specific 13 gene signature provides valuable information for understanding the molecular cause of HIBM and may also pave the way for better diagnosis and effective therapeutic strategies for many muscular disorders.
Collapse
|
41
|
De Somer L, Bader-Meunier B, Breton S, Brachi S, Wouters C, Zulian F. Dry synovitis, a rare entity distinct from juvenile idiopathic arthritis. Pediatr Rheumatol Online J 2023; 21:7. [PMID: 36691078 PMCID: PMC9872413 DOI: 10.1186/s12969-023-00789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/15/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Dry synovitis (DS) is a rare entity as only a few cases have been reported to date. We describe the clinical features, radiological manifestations and course of DS in comparison with rheumatoid factor negative polyarticular juvenile idiopathic arthritis (RFneg-polyJIA). METHODS We performed a multicenter retrospective collection of data of DS patients who presented with progressive joint limitations without palpable synovitis, absence of elevated acute phase reactants, negative ANA and RF, and imaging showing joint and/or osteochondral involvement. For comparative purposes, we included a cohort of RF neg-polyJIA patients. RESULTS Twelve DS patients, 8F/4 M, with mean age at onset of 6.1 years, were included. Presenting signs comprised delayed motor development, functional limitations and/or progressive stiffness. Clinical examination showed symmetric polyarticular involvement with variable muscular atrophy. MRI showed mild, diffuse synovial involvement, without effusion. With time, signs of progressive osteochondral damage became evident, despite treatment. All patients were treated with low-dose corticosteroids and methotrexate. Anti-TNF agents were prescribed in five. The response was variable with limited joint mobility in 11/12, and need of joint replacement in 2. In comparison with a cohort of RFneg-polyJIA, DS patients presented higher number of joint involved (p = 0.0001) and contractures (p = 0.0001), less swelling (p = 0.0001) and prolonged diagnostic delay (p = 0.0001). CONCLUSION DS represents a unique juvenile-onset arthropathy, distinct from polyarticular JIA. Awareness among pediatricians is essential for early recognition and proper treatment. Further studies, including synovial pathology, immunology and genetics may contribute to a better understanding of this rare disorder of childhood.
Collapse
Affiliation(s)
- Lien De Somer
- Department of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium.
| | - Brigitte Bader-Meunier
- grid.412134.10000 0004 0593 9113Department of Pediatric Hematology-Immunology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Sylvain Breton
- grid.412134.10000 0004 0593 9113Department of Pediatric Radiology, Necker-Enfants Malades University Hospital, Paris, France
| | - Sara Brachi
- grid.5608.b0000 0004 1757 3470Pediatric Rheumatology Unit, Department of Woman and Child Health, University of Padua, Padua, Italy
| | - Carine Wouters
- grid.410569.f0000 0004 0626 3338Department of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium ,grid.412134.10000 0004 0593 9113Department of Pediatric Hematology-Immunology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Francesco Zulian
- grid.5608.b0000 0004 1757 3470Pediatric Rheumatology Unit, Department of Woman and Child Health, University of Padua, Padua, Italy
| |
Collapse
|
42
|
Tanboon J, Nishino I. Autosomal Recessive Limb-Girdle Muscular Dystrophies. CURRENT CLINICAL NEUROLOGY 2023:93-121. [DOI: 10.1007/978-3-031-44009-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Kassardjian C, Liewluck T. Systemic Complications of Muscular Dystrophies. CURRENT CLINICAL NEUROLOGY 2023:269-280. [DOI: 10.1007/978-3-031-44009-0_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
44
|
Younger DS. Childhood muscular dystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:461-496. [PMID: 37562882 DOI: 10.1016/b978-0-323-98818-6.00024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Infancy- and childhood-onset muscular dystrophies are associated with a characteristic distribution and progression of motor dysfunction. The underlying causes of progressive childhood muscular dystrophies are heterogeneous involving diverse genetic pathways and genes that encode proteins of the plasma membrane, extracellular matrix, sarcomere, and nuclear membrane components. The prototypical clinicopathological features in an affected child may be adequate to fully distinguish it from other likely diagnoses based on four common features: (1) weakness and wasting of pelvic-femoral and scapular muscles with involvement of heart muscle; (2) elevation of serum muscle enzymes in particular serum creatine kinase; (3) necrosis and regeneration of myofibers; and (4) molecular neurogenetic assessment particularly utilizing next-generation sequencing of the genome of the likeliest candidates genes in an index case or family proband. A number of different animal models of therapeutic strategies have been developed for gene transfer therapy, but so far these techniques have not yet entered clinical practice. Treatment remains for the most part symptomatic with the goal of ameliorating locomotor and cardiorespiratory manifestations of the disease.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
45
|
Pokrovsky MV, Korokin MV, Krayushkina AM, Zhunusov NS, Lapin KN, Soldatova MO, Kuzmin EA, Gudyrev OS, Kochkarova IS, Deikin AV. CONVENTIONAL APPROACHES TO THE THERAPY OF HEREDITARY MYOPATHIES. PHARMACY & PHARMACOLOGY 2022. [DOI: 10.19163/2307-9266-2022-10-5-416-431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of the work was to analyze the available therapeutic options for the conventional therapy of hereditary myopathies.Materials and methods. When searching for the material for writing a review article, such abstract databases as PubMed and Google Scholar were used. The search was carried out on the publications during the period from 1980 to September 2022. The following words and their combinations were selected as parameters for the literature selection: “myopathy”, “Duchenne”, “myodystrophy”, “metabolic”, “mitochondrial”, “congenital”, “symptoms”, “replacement”, “recombinant”, “corticosteroids”, “vitamins”, “tirasemtiv”, “therapy”, “treatment”, “evidence”, “clinical trials”, “patients”, “dichloracetate”.Results. Congenital myopathies are a heterogeneous group of pathologies that are caused by atrophy and degeneration of muscle fibers due to mutations in genes. Based on a number of clinical and pathogenetic features, hereditary myopathies are divided into: 1) congenital myopathies; 2) muscular dystrophy; 3) mitochondrial and 4) metabolic myopathies. At the same time, treatment approaches vary significantly depending on the type of myopathy and can be based on 1) substitution of the mutant protein; 2) an increase in its expression; 3) stimulation of the internal compensatory pathways expression; 4) restoration of the compounds balance associated with the mutant protein function (for enzymes); 5) impact on the mitochondrial function (with metabolic and mitochondrial myopathies); 6) reduction of inflammation and fibrosis (with muscular dystrophies); as well as 7) an increase in muscle mass and strength. The current review presents current data on each of the listed approaches, as well as specific pharmacological agents with a description of their action mechanisms.Conclusion. Currently, the following pharmacological groups are used or undergoing clinical trials for the treatment of various myopathies types: inotropic, anti-inflammatory and antifibrotic drugs, antimyostatin therapy and the drugs that promote translation through stop codons (applicable for nonsense mutations). In addition, metabolic drugs, metabolic enzyme cofactors, mitochondrial biogenesis stimulators, and antioxidants can be used to treat myopathies. Finally, the recombinant drugs alglucosidase and avalglucosidase have been clinically approved for the replacement therapy of metabolic myopathies (Pompe’s disease).
Collapse
Affiliation(s)
| | | | | | | | - K. N. Lapin
- V.A. Negovsky Research Institute of General Reanimatology, Federal Scientific and Clinical Center for Resuscitation and Rehabilitology
| | | | - E. A. Kuzmin
- Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | |
Collapse
|
46
|
Findlay AR, Weihl CC. Genetic-Based Treatment Strategies for Muscular Dystrophy and Congenital Myopathies. Continuum (Minneap Minn) 2022; 28:1800-1816. [PMID: 36537981 PMCID: PMC10496150 DOI: 10.1212/con.0000000000001203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW This article discusses the foundational concepts of genetic treatment strategies employed in neuromuscular medicine, as well as the importance of genetic testing as a requirement for applying gene-based therapy. RECENT FINDINGS Gene therapies have become a reality for several neuromuscular disorders. Exon-skipping and (in Europe) ribosomal read-through approaches are currently available to a subset of patients with Duchenne muscular dystrophy. Microdystrophin gene replacement has shown promise and is nearing the final stages of clinical trials. Numerous gene-based therapies for other muscular dystrophies and congenital myopathies are progressing toward approval as well. SUMMARY Muscular dystrophies and congenital myopathies are a heterogenous group of hereditary muscle disorders. Confirming a diagnosis with genetic testing is not only critical for guiding management, but also an actual prerequisite for current and future gene therapies. Recessive loss-of-function or dominant haploinsufficiency disorders may be treated with gene replacement strategies, whereas dominant negative and toxic gain-of-function disorders are best addressed with a variety of knockdown approaches. It is important to recognize that many therapeutics are mutation specific and will only benefit a subset of individuals with a specific disease.
Collapse
|
47
|
Colman M, Vroman R, Dhooge T, Malfait Z, Symoens S, Burnyté B, Nampoothiri S, Kariminejad A, Malfait F, Syx D. Kyphoscoliotic Ehlers-Danlos syndrome caused by pathogenic variants in FKBP14: Further insights into the phenotypic spectrum and pathogenic mechanisms. Hum Mutat 2022; 43:1994-2009. [PMID: 36054293 DOI: 10.1002/humu.24456] [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: 02/21/2022] [Revised: 07/12/2022] [Accepted: 08/29/2022] [Indexed: 01/25/2023]
Abstract
The Ehlers-Danlos syndromes (EDS) are a heterogeneous group of heritable connective tissue diseases. The autosomal recessive kyphoscoliotic EDS results from deficiency of either lysyl hydroxylase 1 (encoded by PLOD1), crucial for collagen cross-linking; or the peptidyl-prolyl cis-trans isomerase family FK506-binding protein 22 kDa (FKBP22 encoded by FKBP14), a molecular chaperone of types III, IV, VI, and X collagen. This study reports the clinical manifestations of three probands with homozygous pathogenic FKBP14 variants, including the previously reported c.362dupC; p.(Glu122Argfs*7) variant, a novel missense variant (c.587A>G; p.(Asp196Gly)) and a start codon variant (c.2T>G; p.?). Consistent clinical features in the hitherto reported individuals (n = 40) are kyphoscoliosis, generalized joint hypermobility and congenital muscle hypotonia. Severe vascular complications have been observed in 12.5%. A previously unreported feature is microcornea observed in two probands reported here. Both the c.587A>G and the c.362dupC variant cause complete loss of FKBP22. With immunocytochemistry on dermal fibroblasts, we provide the first evidence for intracellular retention of types III and VI collagen in EDS-FKBP14. Scratch wound assays were largely normal. Western blot of proteins involved in the unfolded protein response and autophagy did not reveal significant upregulation in dermal fibroblasts.
Collapse
Affiliation(s)
- Marlies Colman
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Robin Vroman
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Tibbe Dhooge
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Zoë Malfait
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Sofie Symoens
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Biruté Burnyté
- Center for Medical Genetics, Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, Kerala, India
| | | | - Fransiska Malfait
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - Delfien Syx
- Department of Biomolecular Medicine, Center for Medical Genetics, Ghent University, Ghent, Belgium
| |
Collapse
|
48
|
Schüler SC, Liu Y, Dumontier S, Grandbois M, Le Moal E, Cornelison DDW, Bentzinger CF. Extracellular matrix: Brick and mortar in the skeletal muscle stem cell niche. Front Cell Dev Biol 2022; 10:1056523. [PMID: 36523505 PMCID: PMC9745096 DOI: 10.3389/fcell.2022.1056523] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix (ECM) is an interconnected macromolecular scaffold occupying the space between cells. Amongst other functions, the ECM provides structural support to tissues and serves as a microenvironmental niche that conveys regulatory signals to cells. Cell-matrix adhesions, which link the ECM to the cytoskeleton, are dynamic multi-protein complexes containing surface receptors and intracellular effectors that control various downstream pathways. In skeletal muscle, the most abundant tissue of the body, each individual muscle fiber and its associated muscle stem cells (MuSCs) are surrounded by a layer of ECM referred to as the basal lamina. The core scaffold of the basal lamina consists of self-assembling polymeric laminins and a network of collagens that tether proteoglycans, which provide lateral crosslinking, establish collateral associations with cell surface receptors, and serve as a sink and reservoir for growth factors. Skeletal muscle also contains the fibrillar collagenous interstitial ECM that plays an important role in determining tissue elasticity, connects the basal laminae to each other, and contains matrix secreting mesenchymal fibroblast-like cell types and blood vessels. During skeletal muscle regeneration fibroblast-like cell populations expand and contribute to the transitional fibronectin-rich regenerative matrix that instructs angiogenesis and MuSC function. Here, we provide a comprehensive overview of the role of the skeletal muscle ECM in health and disease and outline its role in orchestrating tissue regeneration and MuSC function.
Collapse
Affiliation(s)
- Svenja C. Schüler
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Yuguo Liu
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Grandbois
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Emmeran Le Moal
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - DDW Cornelison
- Division of Biological Sciences Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - C. Florian Bentzinger
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
49
|
Abstract
Muscular dystrophies are a group of genetic disorders characterized by varying degrees of progressive muscle weakness and degeneration. They are clinically and genetically heterogeneous but share the common histological features of dystrophic muscle. There is currently no cure for muscular dystrophies, which is of particular concern for the more disabling and/or lethal forms of the disease. Through the years, several therapies have encouragingly been developed for muscular dystrophies and include genetic, cellular, and pharmacological approaches. In this chapter, we undertake a comprehensive exploration of muscular dystrophy therapeutics under current development. Our review includes antisense therapy, CRISPR, gene replacement, cell therapy, nonsense suppression, and disease-modifying small molecule compounds.
Collapse
|
50
|
Aguti S, Guirguis F, Bönnemann C, Muntoni F, Bolduc V, Zhou H. Exon-Skipping for a Pathogenic COL6A1 Variant in Ullrich Congenital Muscular Dystrophy. Methods Mol Biol 2022; 2587:387-407. [PMID: 36401040 DOI: 10.1007/978-1-0716-2772-3_20] [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] [Indexed: 11/19/2022]
Abstract
Single nucleotide variants that alter splice sites or splicing regulatory elements can lead to the skipping of exons, retention of introns, or insertion of pseudo-exons (PE) into the mature mRNA transcripts. When translated, these changes can disrupt the function of the synthesized protein. Splice-switching antisense oligonucleotides (ASOs) are synthetic, modified nucleic acids that can correct these aberrant splicing events. They are currently in active clinical development for a number of conditions and have been approved by regulatory agencies for the treatment of neuromuscular disorders such as Duchenne muscular dystrophy and spinal muscular atrophy. We have previously reported that splice-switching ASOs effectively skip a pathogenic PE that causes Ullrich congenital muscular dystrophy (UCMD). This erroneous PE insertion is caused by a deep-intronic variant located within intron 11 of COL6A1 (c.930+189 C>T). Here, we describe the detailed protocols and workflow that our labs have used to assess the efficacy of ASOs to skip this PE in vitro. The protocols include designing ASOs; isolating, culturing, and transfecting fibroblasts; extracting RNA and protein; and validating splicing correction at the mRNA and protein levels using quantitative reverse transcription PCR (qRT-PCR) and western blot assays, respectively.
Collapse
Affiliation(s)
- Sara Aguti
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Fady Guirguis
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.,Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Carsten Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Haiyan Zhou
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK. .,Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.
| |
Collapse
|