1
|
Hinkley L, Orbach R, Park J, Alvarez R, Dziewczapolski G, Bönnemann CG, Foley AR. An International Retrospective Early Natural History Study of LAMA2-Related Dystrophies. J Neuromuscul Dis 2024:JND240048. [PMID: 39177609 DOI: 10.3233/jnd-240048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Background LAMA2-related dystrophies (LAMA2-RDs) represent one of the most common forms of congenital muscular dystrophy and have historically been classified into two subtypes: complete or partial deficiency of laminin-211 (merosin). Patients with LAMA2-RD with the typical congenital phenotype manifest severe muscle weakness, delayed motor milestones, joint contractures, failure to thrive, and progressive respiratory insufficiency. Objective While a comprehensive prospective natural history study has been performed in LAMA2-RD patients over 5 years of age, the early natural history of patients with LAMA2-RD 5 years and younger has not been comprehensively characterized. Methods We extracted retrospective data for patients with LAMA2-RD ages birth through 5 years via the Congenital Muscle Disease International Registry (CMDIR). We analyzed the data using a phenotypic classification based on maximal motor milestones to divide patients into two phenotypic groups: "Sit" for those patients who attained that ability to remain seated and "Walk" for those patients who attained the ability to walk independently by 3.5 years of age. Results Sixty patients with LAMA2-RD from 10 countries fulfilled the inclusion criteria. Twenty-four patients had initiated non-invasive ventilation by age 5 years. Hospitalizations during the first years of life were often related to respiratory insufficiency. Feeding/nutritional difficulties and orthopedic issues were commonly reported. Significant elevations of creatine kinase (CK) observed during the neonatal period declined rapidly within the first few months of life. Conclusions This is the largest international retrospective early natural history study of LAMA2-RD to date, contributing essential data for understanding early clinical findings in LAMA2-RD which, along with the data being collected in international, prospective early natural history studies, will help to establish clinical trial readiness. Our proposed nomenclature of LAMA2-RD1 for patients who attain the ability to sit (remain seated) and LAMA2-RD2 for patients who attain the ability to walk independently is aimed at further improving LAMA2-RD classification.
Collapse
Affiliation(s)
- Lauren Hinkley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Rotem Orbach
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Justin Park
- Cure CMD, Congenital Muscle Disease International Registry
| | - Rachel Alvarez
- Cure CMD, Congenital Muscle Disease International Registry
| | | | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Jaramillo CJ, Gibbons T, Rose R, Walsh J. Fatal Case of Exercise Collapse Associated with Sickle Cell Trait with Novel Underlying LAMA2 Mutation. Am J Forensic Med Pathol 2024; 45:177-181. [PMID: 38261548 DOI: 10.1097/paf.0000000000000916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
ABSTRACT Sickle cell trait is typically thought to be an asymptomatic carrier state, but it is rarely associated with exertional rhabdomyolysis in cases termed Exercise Collapse Associated with Sickle Cell Trait (ECAST). In a subset of these cases, underlying disease contributes to the development and/or severity of the ensuing medical complications. We describe the first ever case of ECAST reported in a previously asymptomatic, multiply deployed, highly physically active service member with an underlying heterozygous LAMA2 mutation. Moreover, the mutation identified via whole exome sequencing is a novel, likely pathogenic variant that has yet to be described in the literature.
Collapse
Affiliation(s)
| | - Thomas Gibbons
- 59th Medical Wing, Clinical Investigations and Research Support Laboratory, Joint Base San Antonio-Lackland, San Antonio, TX
| | - Rachel Rose
- Department of Pathology, Womack Army Medical Center, Fort Liberty, NC
| | - John Walsh
- Forensic Pathology Investigations, Armed Forces Medical Examiner System, Dover Air Force Base, DE
| |
Collapse
|
3
|
Noro J, Vilaça-Faria H, Reis RL, Pirraco RP. Extracellular matrix-derived materials for tissue engineering and regenerative medicine: A journey from isolation to characterization and application. Bioact Mater 2024; 34:494-519. [PMID: 38298755 PMCID: PMC10827697 DOI: 10.1016/j.bioactmat.2024.01.004] [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: 10/10/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Biomaterial choice is an essential step during the development tissue engineering and regenerative medicine (TERM) applications. The selected biomaterial must present properties allowing the physiological-like recapitulation of several processes that lead to the reestablishment of homeostatic tissue or organ function. Biomaterials derived from the extracellular matrix (ECM) present many such properties and their use in the field has been steadily increasing. Considering this growing importance, it becomes imperative to provide a comprehensive overview of ECM biomaterials, encompassing their sourcing, processing, and integration into TERM applications. This review compiles the main strategies used to isolate and process ECM-derived biomaterials as well as different techniques used for its characterization, namely biochemical and chemical, physical, morphological, and biological. Lastly, some of their applications in the TERM field are explored and discussed.
Collapse
Affiliation(s)
- Jennifer Noro
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Helena Vilaça-Faria
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
4
|
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
|
5
|
Mastrapasqua M, Rossi R, De Cosmo L, Resta A, Errede M, Bizzoca A, Zampatti S, Resta N, Giardina E, Ruggieri M, Virgintino D, Annese T, Laforgia N, Girolamo F. Autophagy increase in Merosin-Deficient Congenital Muscular Dystrophy type 1A. Eur J Transl Myol 2023; 33:11501. [PMID: 37522802 PMCID: PMC10583158 DOI: 10.4081/ejtm.2023.11501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 08/01/2023] Open
Abstract
The autophagy process recycles dysfunctional cellular components and protein aggregates by sequestering them in autophagosomes directed to lysosomes for enzymatic degradation. A basal level of autophagy is essential for skeletal muscle maintenance. Increased autophagy occurs in several forms of muscular dystrophy and in the merosin-deficient congenital muscular dystrophy 1A mouse model (dy3k/dy3k) lacking the laminin-α2 chain. This pilot study aimed to compare autophagy marker expression and autophagosomes presence using light and electron microscopes and western blotting in diagnostic muscle biopsies from newborns affected by different congenital muscular myopathies and dystrophies. Morphological examination showed dystrophic muscle features, predominance of type 2A myofibers, accumulation of autophagosomes in the subsarcolemmal areas, increased number of autophagosomes overexpressing LC3b, Beclin-1 and ATG5, in the merosin-deficient newborn suggesting an increased autophagy. In Duchenne muscular dystrophy, nemaline myopathy, and spinal muscular atrophy the predominant accumulation of p62+ puncta rather suggests an autophagy impairment.
Collapse
Affiliation(s)
- Mariangela Mastrapasqua
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari 'Aldo Moro', Bari.
| | - Roberta Rossi
- Section of Pathology, Department of Precision and Regenerative Medicine and Jonian Area (DiMePRe-J), University of Bari "Aldo Moro", Bari.
| | - Lucrezia De Cosmo
- Neonatology and Neonatal Intensive Care Unit, Ospedale SS. Annunziata, Taranto.
| | - Annalisa Resta
- Neonatology and Neonatal Intensive Care Unit, Ospedale Miulli, Acquaviva delle Fonti.
| | - Mariella Errede
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari 'Aldo Moro', Bari.
| | - Antonella Bizzoca
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari 'Aldo Moro', Bari.
| | - Stefania Zampatti
- Laboratory of Genomic Medicine - Santa Lucia Foundation - IRCCS, Roma.
| | - Nicoletta Resta
- Medical Genetics Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari "Aldo Moro", Bari.
| | - Emiliano Giardina
- Laboratory of Genomic Medicine - Santa Lucia Foundation - IRCCS, Roma.
| | - Maddalena Ruggieri
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari 'Aldo Moro', Bari.
| | - Daniela Virgintino
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari 'Aldo Moro', Bari.
| | - Tiziana Annese
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari 'Aldo Moro', Bari, Italy; Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari.
| | - Nicola Laforgia
- Neonatology and Neonatal Intensive Care Unit, Department of Interdisciplinary Medicine (DIM), University of Bari "Aldo Moro", Bari.
| | - Francesco Girolamo
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari 'Aldo Moro', Bari.
| |
Collapse
|
6
|
Xu Y, Zhu L, Qian Y, Dong M. Limb girdle muscular dystrophy 23 caused by compound heterozygous mutations of LAMA2 gene. Front Pediatr 2023; 11:1191068. [PMID: 37404563 PMCID: PMC10316388 DOI: 10.3389/fped.2023.1191068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/05/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction Mutations of LAMA2 gene are associated with congenital muscular dystrophy (CMD). The LAMA2-related CMD mainly consists of two diseases, merosin deficient congenital muscular dystrophies type 1A (MDC1A) and limb girdle muscular dystrophy 23 (LGMD23). LGMD23 is characterized by slowly progressive proximal muscle weakness, which primarily affects the lower limbs and results in gait difficulties. Additional clinical features include increased serum creatine kinase, abnormal electromyography with or without white matter abnormalities on brain imaging. Methods Clinical data were collected from a Chinese Han family. Whole-exome sequencing, Sanger sequencing, RT-PCR and TA clone sequencing were performed on the family members. Results Compound heterozygous mutations of LAMA2: c.1693C > T (p. Q565*) (maternally inherited) and c.9212-6T > G (paternally inherited) were identified and confirmed in the proband. The mutation c.1693C > T (p. Q565*) was classified as pathogenic according to American College of Medical Genetics and Genomics (ACMG) guidelines. By performing RT-PCR and TA clone sequencing, an insertion of 40-bp intronic sequence (intron 64) was found in the transcripts of the proband and her father, which resulted in a frameshift and premature truncation codon of the LAMA2. In particular, the variant truncated the LamG domain of the LAMA2. Therefore, the c.9212-6T>G was classified as likely pathogenic according to American College of Medical Genetics and Genomics (ACMG) guidelines. Discussion Our findings described two novel mutations in a girl with LGMDR23, which contributes to the genetic counseling of the family and expands the clinical and molecular spectrums of the rare disease.
Collapse
Affiliation(s)
- Yuqing Xu
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, China
| | - Linyan Zhu
- Department of Obstetrics and Gynaecology, Ningbo First Hospital, Ningbo, China
| | - Yeqing Qian
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, China
| | - Minyue Dong
- Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics, Ministry of Education (Zhejiang University), Hangzhou, China
| |
Collapse
|
7
|
Tran VK, Nguyen NL, Tran LNT, Le PT, Tran AH, Pham TLA, Lien NTK, Xuan NT, Thanh LT, Ta TV, Tran TH, Nguyen HH. Merosin-deficient congenital muscular dystrophy type 1a: detection of LAMA2 variants in Vietnamese patients. Front Genet 2023; 14:1183663. [PMID: 37388928 PMCID: PMC10301838 DOI: 10.3389/fgene.2023.1183663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023] Open
Abstract
Background: Merosin-deficient congenital muscular dystrophy type 1A (MDC1A), also known as laminin-α2 chain-deficient congenital muscular dystrophy (LAMA2-MD), is an autosomal recessive disease caused by biallelic variants in the LAMA2 gene. In MDC1A, laminin- α2 chain expression is absent or significantly reduced, leading to some early-onset clinical symptoms including severe hypotonia, muscle weakness, skeletal deformity, non-ambulation, and respiratory insufficiency. Methods: Six patients from five unrelated Vietnamese families presenting with congenital muscular dystrophy were investigated. Targeted sequencing was performed in the five probands. Sanger sequencing was carried out in their families. Multiplex ligation-dependent probe amplification was performed in one family to examine an exon deletion. Results: Seven variants of the LAMA2 (NM_000426) gene were identified and classified as pathogenic/likely pathogenic variants using American College of Medical Genetics and Genomics criteria. Two of these variants were not reported in the literature, including c.7156-5_7157delinsT and c.8974_8975insTGAT. Sanger sequencing indicated their parents as carriers. The mothers of family 4 and family 5 were pregnant and a prenatal testing was performed. The results showed that the fetus of the family 4 only carries c.4717 + 5G>A in the heterozygous form, while the fetus of the family 5 carries compound heterozygous variants, including a deletion of exon 3 and c.4644C>A. Conclusion: Our findings not only identified the underlying genetic etiology for the patients, but also provided genetic counseling for the parents whenever they have an offspring.
Collapse
Affiliation(s)
- Van Khanh Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Ngoc-Lan Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Lan Ngoc Thi Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Phuong Thi Le
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Anh Hai Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Tuan L. A. Pham
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Nguyen Thi Kim Lien
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Nguyen Thi Xuan
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Le Tat Thanh
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Thanh Van Ta
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Thinh Huy Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Huy-Hoang Nguyen
- Institute of Genome Research, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
8
|
Oliveira-Santos A, Dagda M, Wittmann J, Smalley R, Burkin DJ. Vemurafenib improves muscle histopathology in a mouse model of LAMA2-related congenital muscular dystrophy. Dis Model Mech 2023; 16:dmm049916. [PMID: 37021539 PMCID: PMC10184677 DOI: 10.1242/dmm.049916] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Laminin-α2-related congenital muscular dystrophy (LAMA2-CMD) is a neuromuscular disease affecting around 1-9 in 1,000,000 children. LAMA2-CMD is caused by mutations in the LAMA2 gene resulting in the loss of laminin-211/221 heterotrimers in skeletal muscle. LAMA2-CMD patients exhibit severe hypotonia and progressive muscle weakness. Currently, there is no effective treatment for LAMA2-CMD and patients die prematurely. The loss of laminin-α2 results in muscle degeneration, defective muscle repair and dysregulation of multiple signaling pathways. Signaling pathways that regulate muscle metabolism, survival and fibrosis have been shown to be dysregulated in LAMA2-CMD. As vemurafenib is a US Food and Drug Administration (FDA)-approved serine/threonine kinase inhibitor, we investigated whether vemurafenib could restore some of the serine/threonine kinase-related signaling pathways and prevent disease progression in the dyW-/- mouse model of LAMA2-CMD. Our results show that vemurafenib reduced muscle fibrosis, increased myofiber size and reduced the percentage of fibers with centrally located nuclei in dyW-/- mouse hindlimbs. These studies show that treatment with vemurafenib restored the TGF-β/SMAD3 and mTORC1/p70S6K signaling pathways in skeletal muscle. Together, our results indicate that vemurafenib partially improves histopathology but does not improve muscle function in a mouse model of LAMA2-CMD.
Collapse
Affiliation(s)
- Ariany Oliveira-Santos
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Marisela Dagda
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Jennifer Wittmann
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Robert Smalley
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| | - Dean J. Burkin
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno, NV 89557, USA
| |
Collapse
|
9
|
Camelo CG, Artilheiro MC, Martins Moreno CA, Ferraciolli SF, Serafim Silva AM, Fernandes TR, Lucato LT, Rocha AJ, Reed UC, Zanoteli E. Brain MRI Abnormalities, Epilepsy and Intellectual Disability in LAMA2 Related Dystrophy - a Genotype/Phenotype Correlation. J Neuromuscul Dis 2023:JND221638. [PMID: 37182895 DOI: 10.3233/jnd-221638] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND LAMA2-related muscular dystrophy is a disorder that causes muscle weakness and varies in severity, from a severe, congenital type to a milder, late-onset form. However, the disease does not only affect the muscles, but has systemic involvement and can lead to alterations such as brain malformation, epilepsy and intellectual disability. OBJECTIVE Describe the frequency of cortical malformations, epilepsy and intellectual disability in LAMA2-RD in a Brazilian cohort and correlate the neurological findings to genetic and motor function. METHODS This is an observational study of 52 LAMA2-RD patients, who were divided into motor function subgroups and compared based on brain MRI findings, epilepsy, intellectual disability, and type of variants and variant domains. RESULTS 44 patients (84.6%) were only able to sit, and 8 patients (15.4%) were able to walk. 10 patients (19.2%) presented with cortical malformations (polymicrogyria, lissencephaly-pachygyria, and cobblestone),10 patients (19.2%) presented with epilepsy, and 8 (15.4%) had intellectual disability. CNS manifestations correlated with a more severe motor phenotype and none of the patients able to walk presented with cortical malformation or epilepsy. There was a relation between gene variants affecting the laminin-α2 LG-domain and the presence of brain malformation (P = 0.016). There was also a relation between the presence of null variants and central nervous system involvement. A new brazilian possible founder variant was found in 11 patients (21,15%) (c.1255del; p. Ile419Leufs *4). CONCLUSION Cortical malformations, epilepsy and intellectual disability are more frequent among LAMA2-RD patients than previously reported and correlate with motor function severity and the presence of variants affecting the laminin-α2 LG domain. This brings more insight fore phenotype-genotype correlations, shows the importance of reviewing the brain MRI of patients with LAMA2-RD and allows greater attention to the risk of brain malformation, epilepsy, and intellectual disability in those patients with variants that affect the LG domain.
Collapse
Affiliation(s)
- Clara Gontijo Camelo
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | | | | | - Sueli Fazio Ferraciolli
- Department of Radiology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - André Macedo Serafim Silva
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Tatiana Ribeiro Fernandes
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Leandro Tavares Lucato
- Department of Radiology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Antônio José Rocha
- Department of Radiology, Faculdade de Medicina da Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Umbertina Conti Reed
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Edmar Zanoteli
- Department of Neurology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| |
Collapse
|
10
|
Tesoriero C, Greco F, Cannone E, Ghirotto F, Facchinello N, Schiavone M, Vettori A. Modeling Human Muscular Dystrophies in Zebrafish: Mutant Lines, Transgenic Fluorescent Biosensors, and Phenotyping Assays. Int J Mol Sci 2023; 24:8314. [PMID: 37176020 PMCID: PMC10179009 DOI: 10.3390/ijms24098314] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of myopathies characterized by progressive muscle weakness leading to death from heart or respiratory failure. MDs are caused by mutations in genes involved in both the development and organization of muscle fibers. Several animal models harboring mutations in MD-associated genes have been developed so far. Together with rodents, the zebrafish is one of the most popular animal models used to reproduce MDs because of the high level of sequence homology with the human genome and its genetic manipulability. This review describes the most important zebrafish mutant models of MD and the most advanced tools used to generate and characterize all these valuable transgenic lines. Zebrafish models of MDs have been generated by introducing mutations to muscle-specific genes with different genetic techniques, such as (i) N-ethyl-N-nitrosourea (ENU) treatment, (ii) the injection of specific morpholino, (iii) tol2-based transgenesis, (iv) TALEN, (v) and CRISPR/Cas9 technology. All these models are extensively used either to study muscle development and function or understand the pathogenetic mechanisms of MDs. Several tools have also been developed to characterize these zebrafish models by checking (i) motor behavior, (ii) muscle fiber structure, (iii) oxidative stress, and (iv) mitochondrial function and dynamics. Further, living biosensor models, based on the expression of fluorescent reporter proteins under the control of muscle-specific promoters or responsive elements, have been revealed to be powerful tools to follow molecular dynamics at the level of a single muscle fiber. Thus, zebrafish models of MDs can also be a powerful tool to search for new drugs or gene therapies able to block or slow down disease progression.
Collapse
Affiliation(s)
- Chiara Tesoriero
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Francesca Greco
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Elena Cannone
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Francesco Ghirotto
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Nicola Facchinello
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
| | - Marco Schiavone
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Andrea Vettori
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| |
Collapse
|
11
|
Lake NJ, Phua J, Liu W, Moors T, Axon S, Lek M. Estimating the Prevalence of LAMA2 Congenital Muscular Dystrophy using Population Genetic Databases. J Neuromuscul Dis 2023; 10:381-387. [PMID: 37005889 DOI: 10.3233/jnd-221552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Background: Recessive pathogenic variants in LAMA2 resulting in complete or partial loss of laminin α2 protein cause congenital muscular dystrophy (LAMA2 CMD). The prevalence of LAMA2 CMD has been estimated by epidemiological studies to lie between 1.36–20 cases per million. However, prevalence estimates from epidemiological studies are vulnerable to inaccuracies owing to challenges with studying rare diseases. Population genetic databases offer an alternative method for estimating prevalence. Objective: We aim to use population allele frequency data for reported and predicted pathogenic variants to estimate the birth prevalence of LAMA2 CMD. Methods: A list of reported pathogenic LAMA2 variants was compiled from public databases, and supplemented with predicted loss of function (LoF) variants in the Genome Aggregation Database (gnomAD). gnomAD allele frequencies for 273 reported pathogenic and predicted LoF LAMA2 variants were used to calculate disease prevalence using a Bayesian methodology. Results: The world-wide birth prevalence of LAMA2 CMD was estimated to be 8.3 per million (95% confidence interval (CI) 6.27 –10.5 per million). The prevalence estimates for each population in gnomAD varied, ranging from 1.79 per million in East Asians (95% CI 0.63 –3.36) to 10.1 per million in Europeans (95% CI 6.74 –13.9). These estimates were generally consistent with those from epidemiological studies, where available. Conclusions: We provide robust world-wide and population-specific birth prevalence estimates for LAMA2 CMD, including for non-European populations in which LAMA2 CMD prevalence hadn’t been studied. This work will inform the design and prioritization of clinical trials for promising LAMA2 CMD treatments.
Collapse
Affiliation(s)
- Nicole J. Lake
- Yale School of Medicine, New Haven, CT, USA
- Murdoch Children’s Research Institute, Melbourne, VIC, Australia
| | - Joel Phua
- Masters Program in Biotechnology, UCSI University, Kuala Lumpur, Malaysia
| | - Wei Liu
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | | | | | - Monkol Lek
- Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
12
|
Chen Z, Kelly JR, Morales JE, Sun RC, De A, Burkin DJ, McCarty JH. The alpha7 integrin subunit in astrocytes promotes endothelial blood-brain barrier integrity. Development 2023; 150:dev201356. [PMID: 36960827 PMCID: PMC10112902 DOI: 10.1242/dev.201356] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/22/2023] [Indexed: 03/25/2023]
Abstract
The blood-brain barrier (BBB) is a vascular endothelial cell boundary that partitions the circulation from the central nervous system to promote normal brain health. We have a limited understanding of how the BBB is formed during development and maintained in adulthood. We used quantitative transcriptional profiling to investigate whether specific adhesion molecules are involved in BBB functions, with an emphasis on understanding how astrocytes interact with endothelial cells. Our results reveal a striking enrichment of multiple genes encoding laminin subunits as well as the laminin receptor gene Itga7, which encodes the alpha7 integrin subunit, in astrocytes. Genetic ablation of Itga7 in mice led to aberrant BBB permeability and progressive neurological pathologies. Itga7-/- mice also showed a reduction in laminin protein expression in parenchymal basement membranes. Blood vessels in the Itga7-/- brain showed separation from surrounding astrocytes and had reduced expression of the tight junction proteins claudin 5 and ZO-1. We propose that the alpha7 integrin subunit in astrocytes via adhesion to laminins promotes endothelial cell junction integrity, all of which is required to properly form and maintain a functional BBB.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jack R. Kelly
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - John E. Morales
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Raymond C. Sun
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Arpan De
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Dean J. Burkin
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Joseph H. McCarty
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
13
|
Arockiaraj AI, Johnson MA, Munir A, Ekambaram P, Lucas PC, McAllister-Lucas LM, Kemaladewi DU. CRISPRa-induced upregulation of human LAMA1 compensates for LAMA2-deficiency in Merosin-deficient congenital muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531347. [PMID: 36945402 PMCID: PMC10028808 DOI: 10.1101/2023.03.06.531347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Merosin-deficient congenital muscular dystrophy (MDC1A) is an autosomal recessive disorder caused by mutations in the LAMA2 gene, resulting in a defective form of the extracellular matrix protein laminin-α2 (LAMA2). Individuals diagnosed with MDC1A exhibit progressive muscle wasting and declining neuromuscular functions. No treatments for this disorder are currently available. We previously showed that postnatal Lama1 upregulation, achieved through CRISPR activation (CRISPRa), compensates for Lama2 deficiency and prevents neuromuscular pathophysiology in a mouse model of MDC1A. In this study, we assessed the feasibility of upregulating human LAMA1 as a potential therapeutic strategy for individuals with MDC1A, regardless of their mutations. We hypothesized that CRISPRa-mediated upregulation of human LAMA1 would compensate for the lack of LAMA2 and rescue cellular abnormalities in MDC1A fibroblasts. Global transcriptomic and pathway enrichment analyses of fibroblasts collected from individuals carrying pathogenic LAMA2 mutations, compared with healthy controls, indicated higher expression of transcripts encoding proteins that contribute to wound healing, including Transforming Growth Factor-β (TGF-β) and Fibroblast Growth Factor (FGF). These findings were supported by wound-healing assays indicating that MDC1A fibroblasts migrated significantly more rapidly than the controls. Subsequently, we treated the MDC1A fibroblasts with SadCas9-2XVP64 and sgRNAs targeting the LAMA1 promoter. We observed robust LAMA1 expression, which was accompanied by significant decreases in cell migration and expression of FGFR2, TGF-β2, and ACTA2, which are involved in the wound-healing mechanism in MDC1A fibroblasts. Collectively, our data suggest that CRISPRa-mediated LAMA1 upregulation may be a feasible mutation-independent therapeutic approach for MDC1A. This strategy might be adapted to address other neuromuscular diseases and inherited conditions in which strong compensatory mechanisms have been identified.
Collapse
Affiliation(s)
- Annie I. Arockiaraj
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Marie A. Johnson
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Anushe Munir
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Prasanna Ekambaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Peter C. Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Dwi U. Kemaladewi
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
14
|
Wang DZ, Li BH, Ma Q, Yu Z, Chen K, He Y, Tan S. Novel compound heterozygous mutations of LAMA2-limb-girdle muscular dystrophy: A case report and literature review. Front Neurol 2023; 14:1078151. [PMID: 36860576 PMCID: PMC9968920 DOI: 10.3389/fneur.2023.1078151] [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: 10/24/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
The laminin α2 (LAMA2) gene pathogenic variants can lead to limb-girdle muscular dystrophy (known as LGMDR23), which is rarely reported and characterized by proximal weakness in the limbs. We present the case of a 52-year-old woman who gradually developed weakness in both lower extremities since the age of 32 years. Magnetic resonance imaging (MRI) brain showed symmetrical sphenoid wings-like white matter demyelination in bilateral lateral ventricles. Electromyography showed quadriceps muscle damage on the bilateral lower extremity. Next-generation sequencing (NGS) found two loci variations in the LAMA2 gene, i.e., c.2749 + 2dup and c.8689C>T. This case highlights the importance of considering LGMDR23 in patients presenting with weakness and white matter demyelination on MRI brain and further expands the gene variants spectrum of LGMDR23.
Collapse
Affiliation(s)
- Duo-Zi Wang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Bing-Hu Li
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiong Ma
- Department of Neurology, The First People's Hospital of Liangshan in Yi Autonomous Prefecture, Xichang, China
| | - Zhou Yu
- Department of Psychosomatic Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Kai Chen
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying He
- Department of Psychosomatic Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Song Tan
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Chengdu, China,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China,*Correspondence: Song Tan ✉
| |
Collapse
|
15
|
Stojkovic T, Masingue M, Métay C, Romero NB, Eymard B, Ben Yaou R, Rialland L, Drunat S, Gartioux C, Nelson I, Allamand V, Bonne G, Villar-Quiles RN. LAMA2-Related Muscular Dystrophy: The Importance of Accurate Phenotyping and Brain Imaging in the Diagnosis of LGMD. J Neuromuscul Dis 2023; 10:125-133. [PMID: 36373293 DOI: 10.3233/jnd-221555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We report three siblings from a non-consanguineous family presenting with contractural limb-girdle phenotype with intrafamilial variability. Muscle MRI showed posterior thigh and quadriceps involvement with a sandwich-like sign. Whole-exome sequencing identified two compound heterozygous missense TTN variants and one heterozygous LAMA2 variant. Brain MRI performed because of concentration difficulties in one of the siblings evidenced white-matter abnormalities, subsequently found in the others. The genetic analysis was re-oriented, revealing a novel pathogenic intronic LAMA2 variant which confirmed the LAMA2-RD diagnosis. This work highlights the importance of a thorough clinical phenotyping and the importance of brain imaging, in order to orientate and interpret the genetic analysis.
Collapse
Affiliation(s)
- Tanya Stojkovic
- APHP, Reference Center for Neuromuscular Disorders, Institut de Myologie, Pitié-Salpêtrière Hospital, Paris, France.,INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Marion Masingue
- APHP, Reference Center for Neuromuscular Disorders, Institut de Myologie, Pitié-Salpêtrière Hospital, Paris, France
| | - Corinne Métay
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France.,AP-HP, Centre de Génétique Moléculaire et Chromosomique, UF Cardiomyogénétique et Myogénétique Moléculaire et Cellulaire, Pitié-Salpêtrière Hospital, Paris, France
| | - Norma B Romero
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France.,Neuromuscular Morphology Unit, Institut de Myologie, APHP, Pitié-Salpêtrière Hospital, Paris, France
| | - Bruno Eymard
- APHP, Reference Center for Neuromuscular Disorders, Institut de Myologie, Pitié-Salpêtrière Hospital, Paris, France
| | - Rabah Ben Yaou
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Laetitia Rialland
- AP-HP, Centre de Génétique Moléculaire et Chromosomique, UF Cardiomyogénétique et Myogénétique Moléculaire et Cellulaire, Pitié-Salpêtrière Hospital, Paris, France
| | - Séverine Drunat
- Département de génétique, Hôpital Universitaire Robert Debré, Paris, France
| | - Corine Gartioux
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Isabelle Nelson
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Valérie Allamand
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Gisèle Bonne
- INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Rocio Nur Villar-Quiles
- APHP, Reference Center for Neuromuscular Disorders, Institut de Myologie, Pitié-Salpêtrière Hospital, Paris, France.,INSERM, Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Goult BT, von Essen M, Hytönen VP. The mechanical cell - the role of force dependencies in synchronising protein interaction networks. J Cell Sci 2022; 135:283155. [PMID: 36398718 PMCID: PMC9845749 DOI: 10.1242/jcs.259769] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The role of mechanical signals in the proper functioning of organisms is increasingly recognised, and every cell senses physical forces and responds to them. These forces are generated both from outside the cell or via the sophisticated force-generation machinery of the cell, the cytoskeleton. All regions of the cell are connected via mechanical linkages, enabling the whole cell to function as a mechanical system. In this Review, we define some of the key concepts of how this machinery functions, highlighting the critical requirement for mechanosensory proteins, and conceptualise the coupling of mechanical linkages to mechanochemical switches that enables forces to be converted into biological signals. These mechanical couplings provide a mechanism for how mechanical crosstalk might coordinate the entire cell, its neighbours, extending into whole collections of cells, in tissues and in organs, and ultimately in the coordination and operation of entire organisms. Consequently, many diseases manifest through defects in this machinery, which we map onto schematics of the mechanical linkages within a cell. This mapping approach paves the way for the identification of additional linkages between mechanosignalling pathways and so might identify treatments for diseases, where mechanical connections are affected by mutations or where individual force-regulated components are defective.
Collapse
Affiliation(s)
- Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, Kent, UK,Authors for correspondence (; )
| | - Magdaléna von Essen
- Faculty of Medicine and Health Technology, Tampere University, FI-33100 Tampere, Finland
| | - Vesa P. Hytönen
- Faculty of Medicine and Health Technology, Tampere University, FI-33100 Tampere, Finland,Fimlab Laboratories, FI-33520 Tampere, Finland,Authors for correspondence (; )
| |
Collapse
|
19
|
Mianesaz H, Ghalamkari S, Salehi M, Behnam M, Hosseinzadeh M, Basiri K, Ghasemi M, Sedghi M, Ansari B. Causative variants linked with limb girdle muscular dystrophy in an Iranian population: 6 novel variants. Mol Genet Genomic Med 2022; 11:e2101. [PMID: 36374152 PMCID: PMC9938754 DOI: 10.1002/mgg3.2101] [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: 06/12/2022] [Revised: 10/03/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Limb-girdle muscular dystrophy (LGMD) is a non-syndromic muscular dystrophy caused by variations in the genes involved in muscle structure, function and repair. The heterogeneity in the severity, progression, age of onset, and causative genes makes next-generation sequencing (NGS) a necessary approach for the proper diagnosis of LGMD. METHODS In this article, 26 Iranian patients with LGMD criteria were diagnosed with disease variants in the genes encoding calpain3, dysferlin, sarcoglycans and Laminin α-2. Patients were referred to the hospital with variable distribution of muscle wasting and progressive weakness in the body. The symptoms along with biochemical and EMG tests were suggestive of LGMD; thus the genomic DNA of patients were investigated by whole-exome sequencing including flanking intronic regions. The target genes were explored for the disease-causing variants. Moreover, the consequence of the amino acid alterations on proteins' secondary structure and function was investigated for a better understanding of the pathogenicity of variants. Variants were sorted based on the genomic region, type and clinical significance. RESULTS In a comprehensive investigation of previous clinical records, 6 variations were determined as novel, including c.1354-2 A > T and c.3169_3172dupCGGC in DYSF, c.568 G > T in SGCD, c.7243 C > T, c.8662_8663 insT and c. 4397G > C in LAMA2. Some of the detected variants were located in functional domains and/or near to the post-translational modification sites, altering or removing highly conserved regions of amino acid sequence.
Collapse
Affiliation(s)
- Hamidreza Mianesaz
- Department of Human Genetics, Medical SchoolUniversity of DebrecenDebrecenHungary,Department of Genetics and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran
| | - Safoura Ghalamkari
- Department of Genetics and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran,Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Mansoor Salehi
- Department of Genetics and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran,Cellular, Molecular and Genetics Research CenterIsfahan University of Medical SciencesIsfahanIran
| | - Mahdiyeh Behnam
- Cellular, Molecular and Genetics Research CenterIsfahan University of Medical SciencesIsfahanIran,Student Research CommitteeSemnan University of Medical ScienceSemnanIran
| | - Majid Hosseinzadeh
- Department of Genetics and Molecular BiologyIsfahan University of Medical SciencesIsfahanIran,Medical Genetics Laboratory, Alzahra University HospitalIsfahan University of Medical SciencesIsfahanIran
| | - Keivan Basiri
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical ScienceTehranIran,Department of NeurologyIsfahan University of Medical SciencesIsfahanIran
| | - Majid Ghasemi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical ScienceTehranIran,Department of NeurologyIsfahan University of Medical SciencesIsfahanIran
| | - Maryam Sedghi
- Medical Genetics Laboratory, Alzahra University HospitalIsfahan University of Medical SciencesIsfahanIran,Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical ScienceTehranIran
| | - Behnaz Ansari
- Department of NeurologyIsfahan University of Medical SciencesIsfahanIran,Isfahan Neuroscience Research Center, ALzahra Research InstituteIsfahan University of Medical ScienceIsfahanIran
| |
Collapse
|
20
|
Basement Membrane Changes of Myofiber and Fibrosis in Sternocleidomastoid Muscle of Congenital Muscular Torticollis. J Craniofac Surg 2022; 33:2704-2710. [DOI: 10.1097/scs.0000000000008781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
|
21
|
Limbach LE, Penick RL, Casseday RS, Hyland MA, Pontillo EA, Ayele AN, Pitts KM, Ackerman SD, Harty BL, Herbert AL, Monk KR, Petersen SC. Peripheral nerve development in zebrafish requires muscle patterning by tcf15/paraxis. Dev Biol 2022; 490:37-49. [PMID: 35820658 DOI: 10.1016/j.ydbio.2022.07.001] [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: 03/04/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/03/2022]
Abstract
The vertebrate peripheral nervous system (PNS) is an intricate network that conveys sensory and motor information throughout the body. During development, extracellular cues direct the migration of axons and glia through peripheral tissues. Currently, the suite of molecules that govern PNS axon-glial patterning is incompletely understood. To elucidate factors that are critical for peripheral nerve development, we characterized the novel zebrafish mutant, stl159, that exhibits abnormalities in PNS patterning. In these mutants, motor and sensory nerves that develop adjacent to axial muscle fail to extend normally, and neuromasts in the posterior lateral line system, as well as neural crest-derived melanocytes, are incorrectly positioned. The stl159 genetic lesion lies in the basic helix-loop-helix (bHLH) transcription factor tcf15, which has been previously implicated in proper development of axial muscles. We find that targeted loss of tcf15 via CRISPR-Cas9 genome editing results in the PNS patterning abnormalities observed in stl159 mutants. Because tcf15 is expressed in developing muscle prior to nerve extension, rather than in neurons or glia, we predict that tcf15 non-cell-autonomously promotes peripheral nerve patterning in zebrafish through regulation of extracellular patterning cues. Our work underscores the importance of muscle-derived factors in PNS development.
Collapse
Affiliation(s)
| | - Rocky L Penick
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
| | - Rudy S Casseday
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
| | | | | | - Afomia N Ayele
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
| | | | - Sarah D Ackerman
- Department of Developmental Biology, Washington University in St. Louis, MO, USA
| | - Breanne L Harty
- Department of Developmental Biology, Washington University in St. Louis, MO, USA
| | - Amy L Herbert
- Department of Developmental Biology, Washington University in St. Louis, MO, USA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University in St. Louis, MO, USA
| | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, OH, USA; Department of Biology, Kenyon College, Gambier, OH, USA; Department of Developmental Biology, Washington University in St. Louis, MO, USA.
| |
Collapse
|
22
|
Whole exome sequencing identified a novel LAMA2 frameshift variant causing merosin-deficient congenital muscular dystrophy in a patient with cardiomyopathy, and autism-like behaviors. Neuromuscul Disord 2022; 32:776-784. [DOI: 10.1016/j.nmd.2022.07.400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022]
|
23
|
Zou S, Pan BX. Post-synaptic specialization of the neuromuscular junction: junctional folds formation, function, and disorders. Cell Biosci 2022; 12:93. [PMID: 35718785 PMCID: PMC9208267 DOI: 10.1186/s13578-022-00829-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/05/2022] [Indexed: 11/14/2022] Open
Abstract
Post-synaptic specialization is critical to the neurotransmitter release and action potential conduction. The neuromuscular junctions (NMJs) are the synapses between the motor neurons and muscle cells and have a more specialized post-synaptic membrane than synapses in the central nervous system (CNS). The sarcolemma within NMJ folded to form some invagination portions called junctional folds (JFs), and they have important roles in maintaining the post-synaptic membrane structure. The NMJ formation and the acetylcholine receptor (AChR) clustering signal pathway have been extensively studied and reviewed. Although it has been suggested that JFs are related to maintaining the safety factor of neurotransmitter release, the formation mechanism and function of JFs are still unclear. This review will focus on the JFs about evolution, formation, function, and disorders. Anticipate understanding of where they are coming from and where we will study in the future.
Collapse
Affiliation(s)
- Suqi Zou
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China.
- School of Life Sciences, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China.
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China
- School of Life Sciences, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China
| |
Collapse
|
24
|
Smith SJ, Fabian L, Sheikh A, Noche R, Cui X, Moore SA, Dowling JJ. Lysosomes and the pathogenesis of merosin-deficient congenital muscular dystrophy. Hum Mol Genet 2022; 31:733-747. [PMID: 34568901 PMCID: PMC9989739 DOI: 10.1093/hmg/ddab278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/01/2021] [Accepted: 09/14/2021] [Indexed: 11/14/2022] Open
Abstract
Congenital muscular dystrophy type 1A (MDC1A), the most common congenital muscular dystrophy in Western countries, is caused by recessive mutations in LAMA2, the gene encoding laminin alpha 2. Currently, no cure or disease modifying therapy has been successfully developed for MDC1A. Examination of patient muscle biopsies revealed altered distribution of lysosomes. We hypothesized that this redistribution was a novel and potentially druggable aspect of disease pathogenesis. We explored this hypothesis using candyfloss (caf), a zebrafish model of MDC1A. We found that lysosome distribution in caf zebrafish was also abnormal. This altered localization was significantly associated with fiber detachment and could be prevented by blocking myofiber detachment. Overexpression of transcription factor EB, a transcription factor that promotes lysosomal biogenesis, led to increased lysosome content and decreased fiber detachment. We conclude that genetic manipulation of the lysosomal compartment is able to alter the caf zebrafish disease process, suggesting that lysosome function may be a target for disease modification.
Collapse
Affiliation(s)
- Sarah J Smith
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.,Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Family Medicine, University of Calgary, Calgary T2R 0X7, Alberta
| | - Lacramioara Fabian
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Adeel Sheikh
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Ramil Noche
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Zebrafish Genetics and Disease Models Core Facility, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiucheng Cui
- Zebrafish Genetics and Disease Models Core Facility, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Steven A Moore
- Department of Pathology, University of Iowa Medical Center, Iowa City, IA, USA
| | - James J Dowling
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.,Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Division of Neurology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,Department of Paediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
25
|
Boyd A, Montandon M, Wood AJ, Currie PD. FKRP directed fibronectin glycosylation: A novel mechanism giving insights into muscular dystrophies? Bioessays 2022; 44:e2100270. [PMID: 35229908 DOI: 10.1002/bies.202100270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/15/2022]
Abstract
The recently uncovered role of Fukutin-related protein (FKRP) in fibronectin glycosylation has challenged our understanding of the basis of disease pathogenesis in the muscular dystrophies. FKRP is a Golgi-resident glycosyltransferase implicated in a broad spectrum of muscular dystrophy (MD) pathologies that are not fully attributable to the well-described α-Dystroglycan hypoglycosylation. By revealing a new role for FKRP in the glycosylation of fibronectin, a modification critical for the development of the muscle basement membrane (MBM) and its associated muscle linkages, new possibilities for understanding clinical phenotype arise. This modification involves an interaction between FKRP and myosin-10, a protein involved in the Golgi organization and function. These observations suggest a FKRP nexus exists that controls two critical aspects to muscle fibre integrity, both fibre stability at the MBM and its elastic properties. This review explores the new potential disease axis in the context of our current knowledge of muscular dystrophies.
Collapse
Affiliation(s)
- Andrew Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Margo Montandon
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
26
|
Ganassi M, Muntoni F, Zammit PS. Defining and identifying satellite cell-opathies within muscular dystrophies and myopathies. Exp Cell Res 2022; 411:112906. [PMID: 34740639 PMCID: PMC8784828 DOI: 10.1016/j.yexcr.2021.112906] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/12/2021] [Accepted: 10/29/2021] [Indexed: 12/19/2022]
Abstract
Muscular dystrophies and congenital myopathies arise from specific genetic mutations causing skeletal muscle weakness that reduces quality of life. Muscle health relies on resident muscle stem cells called satellite cells, which enable life-course muscle growth, maintenance, repair and regeneration. Such tuned plasticity gradually diminishes in muscle diseases, suggesting compromised satellite cell function. A central issue however, is whether the pathogenic mutation perturbs satellite cell function directly and/or indirectly via an increasingly hostile microenvironment as disease progresses. Here, we explore the effects on satellite cell function of pathogenic mutations in genes (myopathogenes) that associate with muscle disorders, to evaluate clinical and muscle pathological hallmarks that define dysfunctional satellite cells. We deploy transcriptomic analysis and comparison between muscular dystrophies and myopathies to determine the contribution of satellite cell dysfunction using literature, expression dynamics of myopathogenes and their response to the satellite cell regulator PAX7. Our multimodal approach extends current pathological classifications to define Satellite Cell-opathies: muscle disorders in which satellite cell dysfunction contributes to pathology. Primary Satellite Cell-opathies are conditions where mutations in a myopathogene directly affect satellite cell function, such as in Progressive Congenital Myopathy with Scoliosis (MYOSCO) and Carey-Fineman-Ziter Syndrome (CFZS). Primary satellite cell-opathies are generally characterised as being congenital with general hypotonia, and specific involvement of respiratory, trunk and facial muscles, although serum CK levels are usually within the normal range. Secondary Satellite Cell-opathies have mutations in myopathogenes that affect both satellite cells and muscle fibres. Such classification aids diagnosis and predicting probable disease course, as well as informing on treatment and therapeutic development.
Collapse
Affiliation(s)
- Massimo Ganassi
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK.
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, United Kingdom; NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, United Kingdom
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
27
|
Christen M, Indzhova V, Guo LT, Jagannathan V, Leeb T, Shelton GD, Brocal J. LAMA2 Nonsense Variant in an Italian Greyhound with Congenital Muscular Dystrophy. Genes (Basel) 2021; 12:1823. [PMID: 34828429 PMCID: PMC8618982 DOI: 10.3390/genes12111823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/30/2022] Open
Abstract
A 4-month-old, male Italian Greyhound with clinical signs of a neuromuscular disease was investigated. The affected dog presented with an abnormal short-strided gait, generalized muscle atrophy, and poor growth since 2-months of age. Serum biochemistry revealed a marked elevation in creatine kinase activity. Electrodiagnostic testing supported a myopathy. Histopathology of muscle biopsies confirmed a dystrophic phenotype with excessive variability in myofiber size, degenerating fibers, and endomysial fibrosis. A heritable form of congenital muscular dystrophy (CMD) was suspected, and a genetic analysis initiated. We sequenced the genome of the affected dog and compared the data to that of 795 control genomes. This search revealed a private homozygous nonsense variant in LAMA2, XM_022419950.1:c.3285G>A, predicted to truncate 65% of the open reading frame of the wild type laminin α2 protein, XP_022275658.1:p.(Trp1095*). Immunofluorescent staining performed on muscle cryosections from the affected dog confirmed the complete absence of laminin α2 in skeletal muscle. LAMA2 loss of function variants were shown to cause severe laminin α2-related CMD in humans, mouse models, and in one previously described dog. Our data together with current knowledge on other species suggest the LAMA2 nonsense variant as cause for the CMD phenotype in the investigated dog.
Collapse
Affiliation(s)
- Matthias Christen
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (M.C.); (V.J.)
| | - Victoria Indzhova
- Neurology-Neurosurgery Service, Willows Veterinary Centre and Referral Service, Solihull B90 4NH, West Midlands, UK;
| | - Ling T. Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709, USA; (L.T.G.); (G.D.S.)
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (M.C.); (V.J.)
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; (M.C.); (V.J.)
| | - G. Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709, USA; (L.T.G.); (G.D.S.)
| | - Josep Brocal
- Department of Neurology and Neurosurgery, Anderson Moores Veterinary Specialists, Winchester SO21 2LL, Hampshire, UK;
| |
Collapse
|
28
|
Barraza-Flores P, Bukovec KE, Dagda M, Conner BW, Oliveira-Santos A, Grange RW, Burkin DJ. Laminin-111 protein therapy after disease onset slows muscle disease in a mouse model of laminin-α2 related congenital muscular dystrophy. Hum Mol Genet 2021; 29:2162-2170. [PMID: 32472139 DOI: 10.1093/hmg/ddaa104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/03/2020] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
Laminin-α2 related congenital muscular dystrophy (LAMA2-CMD) is a fatal muscle disease caused by mutations in the LAMA2 gene. Laminin-α2 is critical for the formation of laminin-211 and -221 heterotrimers in the muscle basal lamina. LAMA2-CMD patients exhibit hypotonia from birth and progressive muscle loss that results in developmental delay, confinement to a wheelchair, respiratory insufficiency and premature death. There is currently no cure or effective treatment for LAMA2-CMD. Several studies have shown laminin-111 can serve as an effective protein-replacement therapy for LAMA2-CMD. Studies have demonstrated early treatment with laminin-111 protein results in an increase in life expectancy and improvements in muscle pathology and function. Since LAMA2-CMD patients are often diagnosed after advanced disease, it is unclear if laminin-111 protein therapy at an advanced stage of the disease can have beneficial outcomes. In this study, we tested the efficacy of laminin-111 protein therapy after disease onset in a mouse model of LAMA2-CMD. Our results showed laminin-111 treatment after muscle disease onset increased life expectancy, promoted muscle growth and increased muscle stiffness. Together these studies indicate laminin-111 protein therapy either early or late in the disease process could serve as an effective protein replacement therapy for LAMA2-CMD.
Collapse
Affiliation(s)
- Pamela Barraza-Flores
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Katherine E Bukovec
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Marisela Dagda
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Brandon W Conner
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Ariany Oliveira-Santos
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Dean J Burkin
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
29
|
Shaw L, Sugden CJ, Hamill KJ. Laminin Polymerization and Inherited Disease: Lessons From Genetics. Front Genet 2021; 12:707087. [PMID: 34456976 PMCID: PMC8388930 DOI: 10.3389/fgene.2021.707087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/13/2021] [Indexed: 01/13/2023] Open
Abstract
The laminins (LM) are a family of basement membranes glycoproteins with essential structural roles in supporting epithelia, endothelia, nerves and muscle adhesion, and signaling roles in regulating cell migration, proliferation, stem cell maintenance and differentiation. Laminins are obligate heterotrimers comprised of α, β and γ chains that assemble intracellularly. However, extracellularly these heterotrimers then assemble into higher-order networks via interaction between their laminin N-terminal (LN) domains. In vitro protein studies have identified assembly kinetics and the structural motifs involved in binding of adjacent LN domains. The physiological importance of these interactions has been identified through the study of pathogenic point mutations in LN domains that lead to syndromic disorders presenting with phenotypes dependent on which laminin gene is mutated. Genotype-phenotype comparison between knockout and LN domain missense mutations of the same laminin allows inferences to be drawn about the roles of laminin network assembly in terms of tissue function. In this review, we will discuss these comparisons in terms of laminin disorders, and the therapeutic options that understanding these processes have allowed. We will also discuss recent findings of non-laminin mediators of laminin network assembly and their implications in terms of basement membrane structure and function.
Collapse
Affiliation(s)
| | | | - Kevin J. Hamill
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
30
|
Yuan R, Zhang J, Wang Y, Zhu X, Hu S, Zeng J, Liang F, Tang Q, Chen Y, Chen L, Zhu W, Li M, Mo D. Reorganization of chromatin architecture during prenatal development of porcine skeletal muscle. DNA Res 2021; 28:6261936. [PMID: 34009337 PMCID: PMC8154859 DOI: 10.1093/dnares/dsab003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
Abstract
Myofibres (primary and secondary myofibre) are the basic structure of muscle and the determinant of muscle mass. To explore the skeletal muscle developmental processes from primary myofibres to secondary myofibres in pigs, we conducted an integrative three-dimensional structure of genome and transcriptomic characterization of longissimus dorsi muscle of pig from primary myofibre formation stage [embryonic Day 35 (E35)] to secondary myofibre formation stage (E80). In the hierarchical genomic structure, we found that 11.43% of genome switched compartment A/B status, 14.53% of topologically associating domains are changed intradomain interactions (D-scores) and 2,730 genes with differential promoter–enhancer interactions and (or) enhancer activity from E35 to E80. The alterations of genome architecture were found to correlate with expression of genes that play significant roles in neuromuscular junction, embryonic morphogenesis, skeletal muscle development or metabolism, typically, NEFL, MuSK, SLN, Mef2D and GCK. Significantly, Sox6 and MATN2 play important roles in the process of primary to secondary myofibres formation and increase the regulatory potential score and genes expression in it. In brief, we reveal the genomic reorganization from E35 to E80 and construct genome-wide high-resolution interaction maps that provide a resource for studying long-range control of gene expression from E35 to E80.
Collapse
Affiliation(s)
- Renqiang Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.,Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaman Zhang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yujie Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xingxing Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Silu Hu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jianhua Zeng
- Guangdong YIHAO Food Co., Ltd, Guangzhou 510620, China
| | - Feng Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Luxi Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.,Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wei Zhu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
31
|
El Kadiri Y, Ratbi I, Laarabi FZ, Kriouile Y, Sefiani A, Lyahyai J. Identification of a novel LAMA2 c.2217G > A, p.(Trp739*) mutation in a Moroccan patient with congenital muscular dystrophy: a case report. BMC Med Genomics 2021; 14:113. [PMID: 33882917 PMCID: PMC8060993 DOI: 10.1186/s12920-021-00959-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/12/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Merosin-deficient congenital muscular dystrophy type 1A (MDC1A) is a rare autosomal recessive genetic condition caused by deleterious mutations in the LAMA2 gene encoding the laminin-α2 chain. It is the most frequent subtype of congenital muscular dystrophies (CMDs) characterized by total laminin-α2 deficiency with muscle weakness at birth or in the first six months of life. To the best of our knowledge, this study reports the first molecular diagnosis and genetic defect of this heterogeneous form of CMD performed in a Moroccan medical genetic center using next-generation sequencing (NGS). It allows us to expand the mutational spectrum of the LAMA2 gene. CASE PRESENTATION We report the case of a female Moroccan child with clinical and paraclinical features in favor of a CMD. She has global congenital hypotonia with generalized muscle weakness, psychomotor retardation, increased serum creatine kinase, and normal brain scan at the age of six months. Targeted NGS leads to the identification of a novel homozygous nonsense mutation c.2217G > A, p.(Trp739*) in the exon 16 of LAMA2. Sanger sequencing confirmed this mutation in the affected patient and showed that her parents are heterozygous carriers. CONCLUSIONS A modern genetic analysis by NGS improves the genetic diagnosis pathway for adequate genetic counseling of affected families more precisely. An accession number from the National Center for Biotechnology Information (NCBI) ClinVar database was retrieved for this novel LAMA2 mutation.
Collapse
Affiliation(s)
- Youssef El Kadiri
- Centre de Recherche en Génomique des Pathologies Humaines (GENOPATH), Faculté de Médecine et de Pharmacie, Mohammed V University in Rabat, 10100, Rabat, Morocco.
- Département de Génétique Médicale, Institut National d'Hygiène, BP 769 Agdal, 10090, Rabat, Morocco.
| | - Ilham Ratbi
- Centre de Recherche en Génomique des Pathologies Humaines (GENOPATH), Faculté de Médecine et de Pharmacie, Mohammed V University in Rabat, 10100, Rabat, Morocco
| | - Fatima Zahra Laarabi
- Département de Génétique Médicale, Institut National d'Hygiène, BP 769 Agdal, 10090, Rabat, Morocco
| | - Yamna Kriouile
- Unité de Neuropédiatrie et Maladies Neuro-Métaboliques, Service de Pédiatrie 2- Hôpital d'enfants, Rabat, Morocco
| | - Abdelaziz Sefiani
- Centre de Recherche en Génomique des Pathologies Humaines (GENOPATH), Faculté de Médecine et de Pharmacie, Mohammed V University in Rabat, 10100, Rabat, Morocco
- Département de Génétique Médicale, Institut National d'Hygiène, BP 769 Agdal, 10090, Rabat, Morocco
| | - Jaber Lyahyai
- Centre de Recherche en Génomique des Pathologies Humaines (GENOPATH), Faculté de Médecine et de Pharmacie, Mohammed V University in Rabat, 10100, Rabat, Morocco
| |
Collapse
|
32
|
Jayakody H, Zarei S, Nguyen H, Dalton J, Chen K, Hudgins L, Day J, Withrow K, Pandya A, Teasley J, Dobyns WB, Mathews KD, Moore SA. Cobblestone Malformation in LAMA2 Congenital Muscular Dystrophy (MDC1A). J Neuropathol Exp Neurol 2021; 79:998-1010. [PMID: 32827036 PMCID: PMC7445049 DOI: 10.1093/jnen/nlaa062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/09/2020] [Accepted: 06/04/2020] [Indexed: 11/27/2022] Open
Abstract
Congenital muscular dystrophy type 1A (MDC1A) is caused by recessive variants in laminin α2 (LAMA2). Patients have been found to have white matter signal abnormalities on magnetic resonance imaging (MRI) but rarely structural brain abnormalities. We describe the autopsy neuropathology in a 17-year-old with white matter signal abnormalities on brain MRI. Dystrophic pathology was observed in skeletal muscle, and the sural nerve manifested a mild degree of segmental demyelination and remyelination. A diffuse, bilateral cobblestone appearance, and numerous points of fusion between adjacent gyri were apparent on gross examination of the cerebrum. Brain histopathology included focal disruptions of the glia limitans associated with abnormal cerebral cortical lamination or arrested cerebellar granule cell migration. Subcortical nodular heterotopia was present within the cerebellar hemispheres. Sampling of the centrum semiovale revealed no light microscopic evidence of leukoencephalopathy. Three additional MDC1A patients were diagnosed with cobblestone malformation on brain MRI. Unlike the autopsied patient whose brain had a symmetric distribution of cobblestone pathology, the latter patients had asymmetric involvement, most severe in the occipital lobes. These cases demonstrate that cobblestone malformation may be an important manifestation of the brain pathology in MDC1A and can be present even when patients have a structurally normal brain MRI.
Collapse
Affiliation(s)
- Himali Jayakody
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa.,Department of Neurology, The University of Iowa, Iowa City, Iowa
| | - Sanam Zarei
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa.,Department of Neurology, The University of Iowa, Iowa City, Iowa
| | - Huy Nguyen
- Department of Pathology, The University of Iowa, Iowa City, Iowa
| | - Joline Dalton
- The University of Minnesota, Minneapolis, Minnesota.,Department of Neurology, Stanford University, Palo Alto, California
| | - Kelly Chen
- Department of Pediatrics, Stanford University, Palo Alto, California
| | | | - John Day
- The University of Minnesota, Minneapolis, Minnesota
| | - Kara Withrow
- Department of Pediatrics, Virginia Commonwealth University, Richmond, Virginia
| | - Arti Pandya
- Department of Pediatrics, Virginia Commonwealth University, Richmond, Virginia
| | - Jean Teasley
- Department of Pediatrics, Virginia Commonwealth University, Richmond, Virginia
| | - William B Dobyns
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Katherine D Mathews
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa.,Department of Neurology, The University of Iowa, Iowa City, Iowa
| | - Steven A Moore
- Department of Pathology, The University of Iowa, Iowa City, Iowa
| |
Collapse
|
33
|
Khodaenia N, Farjami Z, Ashnaei AH, Ebrahimi N, Chelvarforoosh N, Urtizberea A, Razmara E, Houshmand M. Novel Homozygous Pathogenic Mutations of LAMA 2 Gene in Patients with Congen ital Muscular Dystrophy. IRANIAN JOURNAL OF CHILD NEUROLOGY 2021; 15:101-106. [PMID: 33558818 PMCID: PMC7856435 DOI: 10.22037/ijcn.v15i1.21649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 01/01/2020] [Indexed: 11/18/2022]
Abstract
The laminin α2 subunit is a protein encoded by the laminin α2 gene(LAMA2) which has the role of adhesion (attachment of cells to one another). Genetics consideration showed that mutation in LAMA2 caused a collection of muscle-wasting conditions called muscular dystrophy. This disorder causes disconnection of muscular cells and degeneration of the musculoskeletal system. In this study, we defined the molecular consideration of three patients with laminin α2 deficiency by clinical presentations of congenital muscular dystrophy. In this regard, 65 exons of the LAMA2 gene were amplified by polymerase chain reaction. Moreover, multiple ligation-dependent probe amplification and next generation sequencing (NGS) were carried out for all the patients. Because of NGS negativity, gene sequencing was performed. Results of searching for rearrangements of the LAMA2 gene enabled us to recognize homozygous pathogenic mutations c.2049_c.2050del, c.7156-2A>G, and c,1303C>T. These mutations produce an out-of-frame transcript that will be degraded by nonsense mediated decay. Therefore, we think these changes are pathogenic ones.
Collapse
Affiliation(s)
- Negar Khodaenia
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zahra Farjami
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.,Department of Modern Sciences& Technologies, Medicine Faculty, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hosein Ashnaei
- Department of Modern Sciences& Technologies, Medicine Faculty, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neshat Ebrahimi
- Laboratory of Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Navid Chelvarforoosh
- Department of Agricultural Biotechnology, Science Faculty, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Ehsan Razmara
- Department of Molecular Genetics, Faculty of Biological Science, Tarbiat Modares, Tehran, Iran
| | - Massoud Houshmand
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
34
|
Petrany MJ, Swoboda CO, Sun C, Chetal K, Chen X, Weirauch MT, Salomonis N, Millay DP. Single-nucleus RNA-seq identifies transcriptional heterogeneity in multinucleated skeletal myofibers. Nat Commun 2020; 11:6374. [PMID: 33311464 PMCID: PMC7733460 DOI: 10.1038/s41467-020-20063-w] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
While the majority of cells contain a single nucleus, cell types such as trophoblasts, osteoclasts, and skeletal myofibers require multinucleation. One advantage of multinucleation can be the assignment of distinct functions to different nuclei, but comprehensive interrogation of transcriptional heterogeneity within multinucleated tissues has been challenging due to the presence of a shared cytoplasm. Here, we utilized single-nucleus RNA-sequencing (snRNA-seq) to determine the extent of transcriptional diversity within multinucleated skeletal myofibers. Nuclei from mouse skeletal muscle were profiled across the lifespan, which revealed the presence of distinct myonuclear populations emerging in postnatal development as well as aging muscle. Our datasets also provided a platform for discovery of genes associated with rare specialized regions of the muscle cell, including markers of the myotendinous junction and functionally validated factors expressed at the neuromuscular junction. These findings reveal that myonuclei within syncytial muscle fibers possess distinct transcriptional profiles that regulate muscle biology.
Collapse
Affiliation(s)
- Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
35
|
Muppirala AN, Limbach LE, Bradford EF, Petersen SC. Schwann cell development: From neural crest to myelin sheath. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e398. [PMID: 33145925 DOI: 10.1002/wdev.398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022]
Abstract
Vertebrate nervous system function requires glial cells, including myelinating glia that insulate axons and provide trophic support that allows for efficient signal propagation by neurons. In vertebrate peripheral nervous systems, neural crest-derived glial cells known as Schwann cells (SCs) generate myelin by encompassing and iteratively wrapping membrane around single axon segments. SC gliogenesis and neurogenesis are intimately linked and governed by a complex molecular environment that shapes their developmental trajectory. Changes in this external milieu drive developing SCs through a series of distinct morphological and transcriptional stages from the neural crest to a variety of glial derivatives, including the myelinating sublineage. Cues originate from the extracellular matrix, adjacent axons, and the developing SC basal lamina to trigger intracellular signaling cascades and gene expression changes that specify stages and transitions in SC development. Here, we integrate the findings from in vitro neuron-glia co-culture experiments with in vivo studies investigating SC development, particularly in zebrafish and mouse, to highlight critical factors that specify SC fate. Ultimately, we connect classic biochemical and mutant studies with modern genetic and visualization tools that have elucidated the dynamics of SC development. This article is categorized under: Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Anoohya N Muppirala
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neuroscience, Kenyon College, Gambier, Ohio, USA
| | | | | | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, Ohio, USA.,Department of Biology, Kenyon College, Gambier, Ohio, USA
| |
Collapse
|
36
|
Abstract
Stem cells (SCs) maintain tissue homeostasis and repair wounds. Despite marked variation in tissue architecture and regenerative demands, SCs often follow similar paradigms in communicating with their microenvironmental "niche" to transition between quiescent and regenerative states. Here we use skin epithelium and skeletal muscle-among the most highly-stressed tissues in our body-to highlight similarities and differences in niche constituents and how SCs mediate natural tissue rejuvenation and perform regenerative acts prompted by injuries. We discuss how these communication networks break down during aging and how understanding tissue SCs has led to major advances in regenerative medicine.
Collapse
Affiliation(s)
- Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| | - Helen M Blau
- Baxter Foundation Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
37
|
Cauley ES, Pittman A, Mummidivarpu S, Karimiani EG, Martinez S, Moroni I, Boostani R, Podini D, Mora M, Jamshidi Y, Hoffman EP, Manzini MC. Novel mutation identification and copy number variant detection via exome sequencing in congenital muscular dystrophy. Mol Genet Genomic Med 2020; 8:e1387. [PMID: 32936536 PMCID: PMC7667317 DOI: 10.1002/mgg3.1387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background Congenital muscular dystrophy type 1A (MDC1A), also termed merosin‐deficient congenital muscular dystrophy (CMD), is a severe form of CMD caused by mutations in the laminin α2 gene (LAMA2). Of the more than 300 likely pathogenic variants found in the Leiden Open Variant Database, the majority are truncating mutations leading to complete LAMA2 loss of function, but multiple copy number variants (CNVs) have also been reported with variable frequency. Methods We collected a cohort of individuals diagnosed with likely MDC1A and sought to identify both single nucleotide variants and small and larger CNVs via exome sequencing by extending the analysis of sequencing data to detect splicing changes and CNVs. Results Standard exome analysis identified multiple novel LAMA2 variants in our cohort, but only four cases carried biallelic variants. Since likely truncating LAMA2 variants are often found in heterozygosity without a second allele, we performed additional splicing and CNV analysis on exome data and identified one splice change outside of the canonical sequences and three CNVs, in the remaining four cases. Conclusions Our findings support the expectation that a portion of MDC1A cases may be caused by at least one CNV allele and show how these changes can be effectively identified by additional analysis of existing exome data.
Collapse
Affiliation(s)
- Edmund S Cauley
- Department of Pharmacology and Physiology, Institute for Neuroscience, The George Washington University, Washington, DC, USA
| | - Alan Pittman
- Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom
| | - Swati Mummidivarpu
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY, USA
| | - Ehsan G Karimiani
- Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom.,Innovative Medical Research Center, Islamic Azad University, Mashhad, Iran
| | - Samantha Martinez
- Department of Pharmacology and Physiology, Institute for Neuroscience, The George Washington University, Washington, DC, USA
| | - Isabella Moroni
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Reza Boostani
- Department of Neurology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Daniele Podini
- Department of Forensic Sciences, The George Washington University, Washington, DC, USA
| | - Marina Mora
- Division of Neuromuscular Diseases and Neuroimmunology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Yalda Jamshidi
- Molecular and Clinical Sciences Institute, St. George's, University of London, London, United Kingdom
| | - Eric P Hoffman
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY, USA
| | - M Chiara Manzini
- Department of Pharmacology and Physiology, Institute for Neuroscience, The George Washington University, Washington, DC, USA.,Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
38
|
Zambon AA, Ridout D, Main M, Mein R, Phadke R, Muntoni F, Sarkozy A. LAMA2-related muscular dystrophy: Natural history of a large pediatric cohort. Ann Clin Transl Neurol 2020; 7:1870-1882. [PMID: 32910545 PMCID: PMC7545609 DOI: 10.1002/acn3.51172] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Objective To characterize natural history of Laminin‐α2 related muscular dystrophies (LAMA2‐RD) to help anticipating complications and identifying reliable outcome measures for clinical trial design and powering. Methods We conducted a retrospective, single‐center, cross‐sectional and longitudinal study on 46 LAMA2‐RD pediatric patients (37 families). Patients were seen at the Dubowitz Neuromuscular Centre, London between 1985 and 2019. Data were collected by case note reviews. Time‐to‐event analysis was performed to estimate median age at complications occurrence. Results Forty two patients had complete deficiency of Laminin‐α2 (CD) and four had partial deficiency (PD). Median age at first and last assessment was 2 years and 12.1 years, respectively. Median follow‐up length was 7.8 years (range 0‐18 years). Seven CD patients died at median age 12 years. One CD and two PD subjects achieved independent ambulation. We observed a linear increase in elbow flexor contractures in CD subjects. Thirty‐two CD and one PD patient developed scoliosis, nine underwent spinal surgery. Twenty‐two CD required nocturnal noninvasive ventilation (median age 11.7 years). CD subjects showed a 2.9% linear annual decline in forced vital capacity % predicted. Nineteen CD and one PD patient required gastrostomy insertion for failure to thrive and/or unsafe swallow (median age 10.9 years). Four CD patients had partial seizures. Mild left cardiac ventricular dysfunction and rhythm disturbances were identified in seven CD patients. Interpretation This retrospective longitudinal study provides long‐term natural history of LAMA2‐RD. This will help management and identification of key milestones of disease progression that could be considered for future therapeutic intervention.
Collapse
Affiliation(s)
- Alberto A Zambon
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK.,Neurology Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Deborah Ridout
- Department of Population, Policy and Practice, UCL Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Marion Main
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| | | | - Rahul Phadke
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK
| |
Collapse
|
39
|
Sarkozy A, Foley AR, Zambon AA, Bönnemann CG, Muntoni F. LAMA2-Related Dystrophies: Clinical Phenotypes, Disease Biomarkers, and Clinical Trial Readiness. Front Mol Neurosci 2020; 13:123. [PMID: 32848593 PMCID: PMC7419697 DOI: 10.3389/fnmol.2020.00123] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
Mutations in the LAMA2 gene affect the production of the α2 subunit of laminin-211 (= merosin) and result in either partial or complete laminin-211 deficiency. Complete merosin deficiency is typically associated with a more severe congenital muscular dystrophy (CMD), clinically manifested by hypotonia and weakness at birth, the development of contractures of large joints, and progressive respiratory involvement. Muscle atrophy and severe weakness typically prevent independent ambulation. Partial merosin deficiency is mostly manifested by later onset limb-girdle weakness and joint contractures so that independent ambulation is typically achieved. Collectively, complete and partial merosin deficiency is referred to as LAMA2-related dystrophies (LAMA2-RDs) and represents one of the most common forms of congenital muscular dystrophies worldwide. LAMA2-RDs are classically characterized by both central and peripheral nervous system involvement with abnormal appearing white matter (WM) on brain MRI and dystrophic appearing muscle on muscle biopsy as well as creatine kinase (CK) levels commonly elevated to >1,000 IU/L. Next-generation sequencing (NGS) has greatly improved diagnostic abilities for LAMA2-RD, and the majority of patients with merosin deficiency carry recessive pathogenic variants in the LAMA2 gene. The existence of multiple animal models for LAMA2-RDs has helped to advance our understanding of laminin-211 and has been instrumental in preclinical research progress and translation to clinical trials. The first clinical trial for the LAMA2-RDs was a phase 1 pharmacokinetic and safety study of the anti-apoptotic compound omigapil, based on preclinical studies performed in the dy W/dy W and dy 2J/dy 2J mouse models. This phase 1 study enabled the collection of pulmonary and motor outcome measures and also provided the opportunity for investigating exploratory outcome measures including muscle ultrasound, muscle MRI and serum, and urine biomarker collection. Natural history studies, including a five-year prospective natural history and comparative outcome measures study in patients with LAMA2-RD, have helped to better delineate the natural history and identify viable outcome measures. Plans for further clinical trials for LAMA2-RDs are presently in progress, highlighting the necessity of identifying adequate, disease-relevant biomarkers, capable of reflecting potential therapeutic changes, in addition to refining the clinical outcome measures and time-to-event trajectory analysis of affected patients.
Collapse
Affiliation(s)
- Anna Sarkozy
- Dubowitz Neuromuscular Centre, Institute of Child Health, Great Ormond Street Hospital for Children, London, United Kingdom
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Alberto A Zambon
- Dubowitz Neuromuscular Centre, Institute of Child Health, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Institute of Child Health, Great Ormond Street Hospital for Children, London, United Kingdom.,National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
40
|
Fabian L, Dowling JJ. Zebrafish Models of LAMA2-Related Congenital Muscular Dystrophy (MDC1A). Front Mol Neurosci 2020; 13:122. [PMID: 32742259 PMCID: PMC7364686 DOI: 10.3389/fnmol.2020.00122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/11/2020] [Indexed: 01/28/2023] Open
Abstract
LAMA2-related congenital muscular dystrophy (CMD; LAMA2-MD), also referred to as merosin deficient CMD (MDC1A), is a severe neonatal onset muscle disease caused by recessive mutations in the LAMA2 gene. LAMA2 encodes laminin α2, a subunit of the extracellular matrix (ECM) oligomer laminin 211. There are currently no treatments for MDC1A, and there is an incomplete understanding of disease pathogenesis. Zebrafish, due to their high degree of genetic conservation with humans, large clutch sizes, rapid development, and optical clarity, have emerged as an excellent model system for studying rare Mendelian diseases. They are particularly suitable as a model for muscular dystrophy because they contain at least one orthologue to all major human MD genes, have muscle that is similar to human muscle in structure and function, and manifest obvious and easily measured MD related phenotypes. In this review article, we present the existing zebrafish models of MDC1A, and discuss their contribution to the understanding of MDC1A pathomechanisms and therapy development.
Collapse
Affiliation(s)
- Lacramioara Fabian
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurology, Hospital for Sick Children, Toronto, ON, Canada.,Departments of Pediatrics and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
41
|
Disabled-2: a positive regulator of the early differentiation of myoblasts. Cell Tissue Res 2020; 381:493-508. [PMID: 32607799 PMCID: PMC7431403 DOI: 10.1007/s00441-020-03237-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/05/2020] [Indexed: 11/25/2022]
Abstract
Dab2 is an adaptor protein and a tumor suppressor. Our previous study has found that Dab2 was expressed in early differentiating skeletal muscles in mouse embryos. In this study, we determined the role of Dab2 in the skeletal muscle differentiation using C2C12 myoblasts in vitro and Xenopus laevis embryos in vivo. The expression of Dab2 was increased in C2C12 myoblasts during the formation of myotubes in vitro. Knockdown of Dab2 expression in C2C12 myoblasts resulted in a reduction of myotube formation, whereas the myotube formation was enhanced upon overexpression of Dab2. Re-expression of Dab2 in C2C12 myoblasts with downregulated expression of Dab2 restored their capacity to form myotubes. Microarray profiling and subsequent network analyses on the 155 differentially expressed genes after Dab2 knockdown showed that Mef2c was an important myogenic transcription factor regulated by Dab2 through the p38 MAPK pathway. It was also involved in other pathways that are associated with muscular development and functions. In Xenopus embryos developed in vivo, XDab2 was expressed in the myotome of somites where various myogenic markers were also expressed. Knockdown of XDab2 expression with antisense morpholinos downregulated the expression of myogenic markers in somites. In conclusion, this study is the first to provide solid evidence to show that Dab2 is a positive regulator of the early myoblast differentiation.
Collapse
|
42
|
Jones LK, Lam R, McKee KK, Aleksandrova M, Dowling J, Alexander SI, Mallawaarachchi A, Cottle DL, Short KM, Pais L, Miner JH, Mallett AJ, Simons C, McCarthy H, Yurchenco PD, Smyth IM. A mutation affecting laminin alpha 5 polymerisation gives rise to a syndromic developmental disorder. Development 2020; 147:dev189183. [PMID: 32439764 PMCID: PMC7540250 DOI: 10.1242/dev.189183] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/30/2020] [Indexed: 12/15/2022]
Abstract
Laminin alpha 5 (LAMA5) is a member of a large family of proteins that trimerise and then polymerise to form a central component of all basement membranes. Consequently, the protein plays an instrumental role in shaping the normal development of the kidney, skin, neural tube, lung and limb, and many other organs and tissues. Pathogenic mutations in some laminins have been shown to cause a range of largely syndromic conditions affecting the competency of the basement membranes to which they contribute. We report the identification of a mutation in the polymerisation domain of LAMA5 in a patient with a complex syndromic disease characterised by defects in kidney, craniofacial and limb development, and by a range of other congenital defects. Using CRISPR-generated mouse models and biochemical assays, we demonstrate the pathogenicity of this variant, showing that the change results in a failure of the polymerisation of α/β/γ laminin trimers. Comparing these in vivo phenotypes with those apparent upon gene deletion in mice provides insights into the specific functional importance of laminin polymerisation during development and tissue homeostasis.
Collapse
Affiliation(s)
- Lynelle K Jones
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Rachel Lam
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Karen K McKee
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08901, USA
| | - Maya Aleksandrova
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08901, USA
| | | | - Stephen I Alexander
- Nephrology Department, Centre for Kidney Research, The Children's Hospital at Westmead, Sydney 2145, New South Wales, Australia
| | - Amali Mallawaarachchi
- Department of Medical Genomics, Royal Prince Alfred Hospital; Garvan Institute of Medical Research, Sydney 2010, New South Wales, Australia
| | - Denny L Cottle
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Kieran M Short
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Lynn Pais
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffery H Miner
- Division of Nephrology, Department of Medicine and Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Andrew J Mallett
- Kidney Health Service, Royal Brisbane and Women's Hospital and the Institute for Molecular Bioscience and Faculty of Medicine, The University of Queensland, Brisbane 4072, Queensland, Australia
| | - Cas Simons
- Murdoch Children's Research Institute, The Royal Children's Hospital Melbourne, Melbourne 3052, Victoria, Australia
| | - Hugh McCarthy
- The Sydney Children's Hospitals Network and the Children's Hospital Westmead Clinical School, University of Sydney, Sydney 2145, New South Wales, Australia
| | - Peter D Yurchenco
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08901, USA
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| |
Collapse
|
43
|
Prediction of the Secretome and the Surfaceome: A Strategy to Decipher the Crosstalk between Adipose Tissue and Muscle during Fetal Growth. Int J Mol Sci 2020; 21:ijms21124375. [PMID: 32575512 PMCID: PMC7353064 DOI: 10.3390/ijms21124375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Crosstalk between adipose and muscular tissues is hypothesized to regulate the number of muscular and adipose cells during fetal growth, with post-natal consequences on lean and fat masses. Such crosstalk largely remains, however, to be described. We hypothesized that a characterization of the proteomes of adipose and muscular tissues from bovine fetuses may enhance the understanding of the crosstalk between these tissues through the prediction of their secretomes and surfaceomes. Proteomic experiments have identified 751 and 514 proteins in fetal adipose tissue and muscle. These are mainly involved in the regulation of cell proliferation or differentiation, but also in pathways such as apoptosis, Wnt signalling, or cytokine-mediated signalling. Of the identified proteins, 51 adipokines, 11 myokines, and 37 adipomyokines were predicted, together with 26 adipose and 13 muscular cell surface proteins. Analysis of protein–protein interactions suggested 13 links between secreted and cell surface proteins that may contribute to the adipose–muscular crosstalk. Of these, an interaction between the adipokine plasminogen and the muscular cell surface alpha-enolase may regulate the fetal myogenesis. The in silico secretome and surfaceome analyzed herein exemplify a powerful strategy to enhance the elucidation of the crosstalk between cell types or tissues.
Collapse
|
44
|
Yamamoto-Shimojima K, Ono H, Imaizumi T, Yamamoto T. Novel LAMA2 variants identified in a patient with white matter abnormalities. Hum Genome Var 2020; 7:16. [PMID: 32509318 PMCID: PMC7248065 DOI: 10.1038/s41439-020-0103-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 11/24/2022] Open
Abstract
Comprehensive genomic analysis was performed in a patient with mild psychomotor developmental delay, elevated creatine kinase, and white matter abnormalities. The results revealed biallelic pathogenic variants in the gene related to merosin-deficient congenital muscular dystrophy, NM_000426.3(LAMA2):c.1338_1339del [p.Gly447Phefs*7] and c.2749 + 2dup, which consist of compound heterozygous involvement with predicted loss-of-function and splicing abnormalities.
Collapse
Affiliation(s)
- Keiko Yamamoto-Shimojima
- Japan Society for the Promotion of Science (RPD), Tokyo, Japan
- Institute of Medical Genetics, Tokyo Women’s Medical University, Tokyo, Japan
- Tokyo Women’s Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
| | - Hiroaki Ono
- Department of Pediatrics, Hiroshima Prefectural Hospital, Hiroshima, Japan
| | - Taichi Imaizumi
- Institute of Medical Genetics, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Toshiyuki Yamamoto
- Institute of Medical Genetics, Tokyo Women’s Medical University, Tokyo, Japan
- Tokyo Women’s Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
45
|
Previtali SC, Zambon AA. LAMA2 Neuropathies: Human Findings and Pathomechanisms From Mouse Models. Front Mol Neurosci 2020; 13:60. [PMID: 32390798 PMCID: PMC7190814 DOI: 10.3389/fnmol.2020.00060] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/26/2020] [Indexed: 12/18/2022] Open
Abstract
Merosin deficient Congenital Muscular Dystrophy (MDC1A), or LAMA2-related muscular dystrophy (LAMA2-RD), is a recessive disorder resulting from mutations in the LAMA2 gene, encoding for the alpha-2 chain of laminin-211. The disease is predominantly characterized by progressive muscular dystrophy affecting patient motor function and reducing life expectancy. However, LAMA2-RD also comprises a developmentally-associated dysmyelinating neuropathy that contributes to the disease progression, in addition to brain abnormalities; the latter often underappreciated. In this brief review, we present data supporting the impact of peripheral neuropathy in the LAMA2-RD phenotype, including both mouse models and human studies. We discuss the molecular mechanisms underlying nerve abnormalities and involved in the laminin-211 pathway, which affects axon sorting, ensheathing and myelination. We conclude with some final considerations of consequences on nerve regeneration and potential therapeutic strategies.
Collapse
Affiliation(s)
- Stefano Carlo Previtali
- Neuromuscular Repair Unit, Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.,Department of Neurology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
46
|
Yap L, Tay HG, Nguyen MT, Tjin MS, Tryggvason K. Laminins in Cellular Differentiation. Trends Cell Biol 2019; 29:987-1000. [DOI: 10.1016/j.tcb.2019.10.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/01/2019] [Accepted: 10/04/2019] [Indexed: 12/21/2022]
|
47
|
Mercuri E, Bönnemann CG, Muntoni F. Muscular dystrophies. Lancet 2019; 394:2025-2038. [PMID: 31789220 DOI: 10.1016/s0140-6736(19)32910-1] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/02/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
Muscular dystrophies are primary diseases of muscle due to mutations in more than 40 genes, which result in dystrophic changes on muscle biopsy. Now that most of the genes responsible for these conditions have been identified, it is possible to accurately diagnose them and implement subtype-specific anticipatory care, as complications such as cardiac and respiratory muscle involvement vary greatly. This development and advances in the field of supportive medicine have changed the standard of care, with an overall improvement in the clinical course, survival, and quality of life of affected individuals. The improved understanding of the pathogenesis of these diseases is being used for the development of novel therapies. In the most common form, Duchenne muscular dystrophy, a few personalised therapies have recently achieved conditional approval and many more are at advanced stages of clinical development. In this Seminar, we concentrate on clinical manifestations, molecular pathogenesis, diagnostic strategy, and therapeutic developments for this group of conditions.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Pediatric Neurology Unit, Università Cattolica del Sacro Cuore Roma, Rome, Italy; Nemo Clinical Centre, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
48
|
Hall TE, Wood AJ, Ehrlich O, Li M, Sonntag CS, Cole NJ, Huttner IG, Sztal TE, Currie PD. Cellular rescue in a zebrafish model of congenital muscular dystrophy type 1A. NPJ Regen Med 2019; 4:21. [PMID: 31754462 PMCID: PMC6858319 DOI: 10.1038/s41536-019-0084-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 10/11/2019] [Indexed: 01/11/2023] Open
Abstract
Laminins comprise structural components of basement membranes, critical in the regulation of differentiation, survival and migration of a diverse range of cell types, including skeletal muscle. Mutations in one muscle enriched Laminin isoform, Laminin alpha2 (Lama2), results in the most common form of congenital muscular dystrophy, congenital muscular dystrophy type 1A (MDC1A). However, the exact cellular mechanism by which Laminin loss results in the pathological spectrum associated with MDC1A remains elusive. Here we show, via live tracking of individual muscle fibres, that dystrophic myofibres in the zebrafish model of MDC1A maintain sarcolemmal integrity and undergo dynamic remodelling behaviours post detachment, including focal sarcolemmal reattachment, cell extension and hyper-fusion with surrounding myoblasts. These observations imply the existence of a window of therapeutic opportunity, where detached cells may be “re-functionalised” prior to their delayed entry into the cell death program, a process we show can be achieved by muscle specific or systemic Laminin delivery. We further reveal that Laminin also acts as a pro-regenerative factor that stimulates muscle stem cell-mediated repair in lama2-deficient animals in vivo. The potential multi-mode of action of Laminin replacement therapy suggests it may provide a potent therapeutic axis for the treatment for MDC1A.
Collapse
Affiliation(s)
- T E Hall
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia.,2Institute for Molecular Bioscience, University of Queensland, 306 Carmody Road, St Lucia, 4067 Australia
| | - A J Wood
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - O Ehrlich
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - M Li
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - C S Sonntag
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - N J Cole
- 3Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, New South Wales 2010 Australia.,4Anatomy & Histology, School of Medical Science, Anderson Stuart Building, Eastern Avenue, The University of Sydney, Sydney, New South Wales 2006 Australia
| | - I G Huttner
- 3Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, New South Wales 2010 Australia
| | - T E Sztal
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia.,5Department of Biological Sciences, Monash University, Victoria, 3800 Australia
| | - P D Currie
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia.,6EMBL Australia, Victorian Node, Monash University, Clayton, VIC 3800 Australia
| |
Collapse
|
49
|
Wu S, Yan M, Ge R, Cheng CY. Crosstalk between Sertoli and Germ Cells in Male Fertility. Trends Mol Med 2019; 26:215-231. [PMID: 31727542 DOI: 10.1016/j.molmed.2019.09.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/16/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022]
Abstract
Spermatogenesis is supported by intricate crosstalk between Sertoli cells and germ cells including spermatogonia, spermatocytes, haploid spermatids, and spermatozoa, which takes place in the epithelium of seminiferous tubules. Sertoli cells, also known as 'mother' or 'nurse' cells, provide nutrients, paracrine factors, cytokines, and other biomolecules to support germ cell development. Sertoli cells facilitate the generation of several biologically active peptides, which include F5-, noncollagenous 1 (NC1)-, and laminin globular (LG)3/4/5-peptide, to modulate cellular events across the epithelium. Here, we critically evaluate the involvement of these peptides in facilitating crosstalk between Sertoli and germ cells to support spermatogenesis and thus fertility. Modulating or mimicking the activity of F5-, NC1-, and LG3/4/5-peptide could be used to enhance the transport across the blood-testis barrier (BTB) of contraceptive drugs or to treat male infertility.
Collapse
Affiliation(s)
- Siwen Wu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA
| | - Ming Yan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Renshan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
50
|
Lindstrand A, Eisfeldt J, Pettersson M, Carvalho CMB, Kvarnung M, Grigelioniene G, Anderlid BM, Bjerin O, Gustavsson P, Hammarsjö A, Georgii-Hemming P, Iwarsson E, Johansson-Soller M, Lagerstedt-Robinson K, Lieden A, Magnusson M, Martin M, Malmgren H, Nordenskjöld M, Norling A, Sahlin E, Stranneheim H, Tham E, Wincent J, Ygberg S, Wedell A, Wirta V, Nordgren A, Lundin J, Nilsson D. From cytogenetics to cytogenomics: whole-genome sequencing as a first-line test comprehensively captures the diverse spectrum of disease-causing genetic variation underlying intellectual disability. Genome Med 2019; 11:68. [PMID: 31694722 PMCID: PMC6836550 DOI: 10.1186/s13073-019-0675-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/09/2019] [Indexed: 12/30/2022] Open
Abstract
Background Since different types of genetic variants, from single nucleotide variants (SNVs) to large chromosomal rearrangements, underlie intellectual disability, we evaluated the use of whole-genome sequencing (WGS) rather than chromosomal microarray analysis (CMA) as a first-line genetic diagnostic test. Methods We analyzed three cohorts with short-read WGS: (i) a retrospective cohort with validated copy number variants (CNVs) (cohort 1, n = 68), (ii) individuals referred for monogenic multi-gene panels (cohort 2, n = 156), and (iii) 100 prospective, consecutive cases referred to our center for CMA (cohort 3). Bioinformatic tools developed include FindSV, SVDB, Rhocall, Rhoviz, and vcf2cytosure. Results First, we validated our structural variant (SV)-calling pipeline on cohort 1, consisting of three trisomies and 79 deletions and duplications with a median size of 850 kb (min 500 bp, max 155 Mb). All variants were detected. Second, we utilized the same pipeline in cohort 2 and analyzed with monogenic WGS panels, increasing the diagnostic yield to 8%. Next, cohort 3 was analyzed by both CMA and WGS. The WGS data was processed for large (> 10 kb) SVs genome-wide and for exonic SVs and SNVs in a panel of 887 genes linked to intellectual disability as well as genes matched to patient-specific Human Phenotype Ontology (HPO) phenotypes. This yielded a total of 25 pathogenic variants (SNVs or SVs), of which 12 were detected by CMA as well. We also applied short tandem repeat (STR) expansion detection and discovered one pathologic expansion in ATXN7. Finally, a case of Prader-Willi syndrome with uniparental disomy (UPD) was validated in the WGS data. Important positional information was obtained in all cohorts. Remarkably, 7% of the analyzed cases harbored complex structural variants, as exemplified by a ring chromosome and two duplications found to be an insertional translocation and part of a cryptic unbalanced translocation, respectively. Conclusion The overall diagnostic rate of 27% was more than doubled compared to clinical microarray (12%). Using WGS, we detected a wide range of SVs with high accuracy. Since the WGS data also allowed for analysis of SNVs, UPD, and STRs, it represents a powerful comprehensive genetic test in a clinical diagnostic laboratory setting.
Collapse
Affiliation(s)
- Anna Lindstrand
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden. .,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. .,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Jesper Eisfeldt
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Maria Pettersson
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Claudia M B Carvalho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Malin Kvarnung
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Giedre Grigelioniene
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Olof Bjerin
- The Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Peter Gustavsson
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Hammarsjö
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Erik Iwarsson
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Johansson-Soller
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Lagerstedt-Robinson
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Agne Lieden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Måns Magnusson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.,Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Marcel Martin
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Helena Malmgren
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Nordenskjöld
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ameli Norling
- The Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ellika Sahlin
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Stranneheim
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Emma Tham
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josephine Wincent
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sofia Ygberg
- The Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Wedell
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Valtteri Wirta
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.,Science for Life Laboratory, Department of Microbiology, Tumor and Cell biology, Karolinska Institutet, Stockholm, Sweden
| | - Ann Nordgren
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Lundin
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Daniel Nilsson
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|