598 results on '"Central core disease"'
Search Results
2. Rycal S48168 (ARM210) for RYR1-related myopathies: a phase one, open-label, dose-escalation trialResearch in context
- Author
-
Joshua J. Todd, Tokunbor A. Lawal, Irene C. Chrismer, Angela Kokkinis, Christopher Grunseich, Minal S. Jain, Melissa R. Waite, Victoria Biancavilla, Shavonne Pocock, Kia Brooks, Christopher J. Mendoza, Gina Norato, Ken Cheung, Willa Riekhof, Pooja Varma, Claudia Colina-Prisco, Magalie Emile-Backer, Katherine G. Meilleur, Andrew R. Marks, Yael Webb, Eugene E. Marcantonio, A. Reghan Foley, Carsten G. Bönnemann, and Payam Mohassel
- Subjects
Central core disease ,Congenital myopathies ,Medicine (General) ,R5-920 - Abstract
Summary: Background: RYR1-related myopathies (RYR1-RM) are caused by pathogenic variants in the RYR1 gene which encodes the type 1 ryanodine receptor (RyR1). RyR1 is the sarcoplasmic reticulum (SR) calcium release channel that mediates excitation-contraction coupling in skeletal muscle. RyR1 sub-conductance, SR calcium leak, reduced RyR1 expression, and oxidative stress often contribute to RYR1-RM pathogenesis. Loss of RyR1-calstabin1 association, SR calcium leak, and increased RyR1 open probability were observed in 17 RYR1-RM patient skeletal muscle biopsies and improved following ex vivo treatment with Rycal compounds. Thus, we initiated a first-in-patient trial of Rycal S48168 (ARM210) in ambulatory adults with genetically confirmed RYR1-RM. Methods: Participants received 120 mg (n = 3) or 200 mg (n = 4) S48168 (ARM210) daily for 29 days. The primary endpoint was safety and tolerability. Exploratory endpoints included S48168 (ARM210) pharmacokinetics (PK), target engagement, motor function measure (MFM)-32, hand grip and pinch strength, timed functional tests, PROMIS fatigue scale, semi-quantitative physical exam strength measurements, and oxidative stress biomarkers. The trial was registered with clinicaltrials.gov (NCT04141670) and was conducted at the National Institutes of Health Clinical Center between October 28, 2019 and December 12, 2021. Findings: S48168 (ARM210) was well-tolerated, did not cause any serious adverse events, and exhibited a dose-dependent PK profile. Three of four participants who received the 200 mg/day dose reported improvements in PROMIS-fatigue at 28 days post-dosing, and also demonstrated improved proximal muscle strength on physical examination. Interpretation: S48168 (ARM210) demonstrated favorable safety, tolerability, and PK, in RYR1-RM affected individuals. Most participants who received 200 mg/day S48168 (ARM210) reported decreased fatigue, a key symptom of RYR1-RM. These results set the foundation for a randomized, double-blind, placebo-controlled proof of concept trial to determine efficacy of S48168 (ARM210) in RYR1-RM. Funding: NINDS and NINR Intramural Research Programs, NIH Clinical Center Bench to Bedside Award (2017-551673), ARMGO Pharma Inc., and its development partner Les Laboratoires Servier.
- Published
- 2024
- Full Text
- View/download PDF
3. Rhabdomyosarcoma Associated with Core Myopathy/Malignant Hyperthermia: Combined Effect of Germline Variants in RYR1 and ASPSCR1 May Play a Role.
- Author
-
Andrade, Pamela V., Santos, Joilson M., Teixeira, Anne C. B., Sogari, Vanessa F., Almeida, Michelle S., Callegari, Fabiano M., Krepischi, Ana C. V., Oliveira, Acary S. B., Vainzof, Mariz, and Silva, Helga C. A.
- Subjects
- *
MALIGNANT hyperthermia , *DERMATOMYOSITIS , *SOFT tissue tumors , *DUCHENNE muscular dystrophy , *RHABDOMYOSARCOMA , *GERM cells - Abstract
Rhabdomyosarcomas have been described in association with thyroid disease, dermatomyositis, Duchenne muscular dystrophy, and in muscular dystrophy models but not in patients with ryanodine receptor-1 gene (RYR1) pathogenic variants. We described here an 18-year-old male who reported a cervical nodule. Magnetic resonance images revealed a mass in the ethmoidal sinus corresponding to rhabdomyosarcoma. As his father died from malignant hyperthermia (MH), an in vitro contracture test was conducted and was positive for MH susceptibility. Muscle histopathological analysis in the biopsy showed the presence of cores. Molecular analysis using NGS sequencing identified germline variants in the RYR1 and ASPSCR1 (alveolar soft part sarcoma) genes. This report expands the spectrum of diseases associated with rhabdomyosarcomas and a possible differential diagnosis of soft tissue tumors in patients with RYR1 variants. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. A novel RYR1 variant in an infant with a unique fetal presentation of central core disease.
- Author
-
Baker, Elizabeth K., Al Gharaibeh, Faris N., Bove, Kevin, Calvo‐Garcia, Maria A., Shillington, Amelle, VandenHeuvel, Katherine, and Cortezzo, DonnaMaria E.
- Abstract
Ryanodine receptor type 1‐related disorder (RYR1‐RD) is the most common subgroup of congenital myopathies with a wide phenotypic spectrum ranging from mild hypotonia to lethal fetal akinesia. Genetic testing for myopathies is imperative as the diagnosis informs counseling regarding prognosis and recurrence risk, treatment options, monitoring, and clinical management. However, diagnostic challenges exist as current options are limited to clinical suspicion prompting testing including: single gene sequencing or familial variant testing, multi‐gene panels, exome, genome sequencing, and invasive testing including muscle biopsy. The timing of diagnosis is of great importance due to the association of RYR1‐RD with malignant hyperthermia (MH). MH is a hypermetabolic crisis that occurs secondary to excessive calcium release in muscles, leading to systemic effects that can progress to shock and death if unrecognized. Given the association of MH with pathogenic variants in RYR1, a diagnosis of RYR1‐RD necessitates an awareness of medical team to avoid potentially triggering agents. We describe a case of a unique fetal presentation with bilateral diaphragmatic eventrations who had respiratory failure, dysmorphic facial features, and profound global hypotonia in the neonatal period. The diagnosis was made at several months of age, had direct implications on her clinical care related to anticipated need to long‐term ventilator support, and ultimately death secondary an arrhythmia as a result of suspected MH. Our report reinforces the importance of having high suspicion for a genetic syndrome and pursuing early, rapid exome or genome sequencing as first line testing in critically ill neonatal intensive care unit patients and further evaluating the pathogenicity of a variant of uncertain significance in the setting of a myopathic phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Expanding the clinical-pathological and genetic spectrum of RYR1-related congenital myopathies with cores and minicores: an Italian population study
- Author
-
Aurora Fusto, Denise Cassandrini, Chiara Fiorillo, Valentina Codemo, Guja Astrea, Adele D’Amico, Lorenzo Maggi, Francesca Magri, Marika Pane, Giorgio Tasca, Daniele Sabbatini, Luca Bello, Roberta Battini, Pia Bernasconi, Fabiana Fattori, Enrico Silvio Bertini, Giacomo Comi, Sonia Messina, Tiziana Mongini, Isabella Moroni, Chiara Panicucci, Angela Berardinelli, Alice Donati, Vincenzo Nigro, Antonella Pini, Melania Giannotta, Claudia Dosi, Enzo Ricci, Eugenio Mercuri, Giovanni Minervini, Silvio Tosatto, Filippo Santorelli, Claudio Bruno, and Elena Pegoraro
- Subjects
RYR1-related myopathies ,Central core disease ,Multi-minicore disease ,Genotype–phenotype correlations ,Neuromuscular disorder ,Protein modelling ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Mutations in the RYR1 gene, encoding ryanodine receptor 1 (RyR1), are a well-known cause of Central Core Disease (CCD) and Multi-minicore Disease (MmD). We screened a cohort of 153 patients carrying an histopathological diagnosis of core myopathy (cores and minicores) for RYR1 mutation. At least one RYR1 mutation was identified in 69 of them and these patients were further studied. Clinical and histopathological features were collected. Clinical phenotype was highly heterogeneous ranging from asymptomatic or paucisymptomatic hyperCKemia to severe muscle weakness and skeletal deformity with loss of ambulation. Sixty-eight RYR1 mutations, generally missense, were identified, of which 16 were novel. The combined analysis of the clinical presentation, disease progression and the structural bioinformatic analyses of RYR1 allowed to associate some phenotypes to mutations in specific domains. In addition, this study highlighted the structural bioinformatics potential in the prediction of the pathogenicity of RYR1 mutations. Further improvement in the comprehension of genotype–phenotype relationship of core myopathies can be expected in the next future: the actual lack of the human RyR1 crystal structure paired with the presence of large intrinsically disordered regions in RyR1, and the frequent presence of more than one RYR1 mutation in core myopathy patients, require designing novel investigation strategies to completely address RyR1 mutation effect.
- Published
- 2022
- Full Text
- View/download PDF
6. Neuromuscular Disease
- Author
-
Cravero, Joseph P., Holzman, Robert S., Mancuso, Thomas J., Cravero, Joseph P., and DiNardo, James A.
- Published
- 2021
- Full Text
- View/download PDF
7. A 6-Year-Old Boy with Respiratory and Feeding Difficulties at Birth, Delayed Gross Motor Milestones, and Facial and Proximal Lower Limb Weakness
- Author
-
Ghosh, Partha S., Cai, Chunyu, Zhou, Lan, editor, Burns, Dennis K., editor, and Cai, Chunyu, editor
- Published
- 2020
- Full Text
- View/download PDF
8. Molecular mechanism of the severe MH/CCD mutation Y522S in skeletal ryanodine receptor (RyR1) by cryo-EM.
- Author
-
Iyer, Kavita A., Yifan Hu, Klose, Thomas, Takashi Murayama, and Samsó, Montserrat
- Subjects
- *
RYANODINE receptors , *ION channels , *MALIGNANT hyperthermia , *MOLECULAR switches , *MUSCLE contraction - Abstract
Ryanodine receptors (RyRs) are main regulators of intracellular Ca2+ release and muscle contraction. The Y522S mutation of RyR1 causes central core disease, a weakening myopathy, and malignant hyperthermia, a sudden and potentially fatal response to anesthetics or heat. Y522 is in the core of the N-terminal subdomain C of RyR1 and the mechanism of how this mutation orchestrates malfunction is unpredictable for this 2-MDa ion channel, which has four identical subunits composed of 15 distinct cytoplasmic domains each. We expressed and purified the RyR1 rabbit homolog, Y523S, from HEK293 cells and reconstituted it in nanodiscs under closed and open states. The highresolution cryogenic electron microscopic (cryo-EM) three-dimensional (3D) structures show that the phenyl ring of Tyr functions in a manner analogous to a “spacer” within an α-helical bundle. Mutation to the much smaller Ser alters the hydrophobic network within the bundle, triggering rearrangement of its α-helices with repercussions in the orientation of most cytoplasmic domains. Examining the mutation-induced readjustments exposed a series of connected α-helices acting as an ∼100 Å-long lever: One end protrudes toward the dihydropyridine receptor, its molecular activator (akin to an antenna), while the other end reaches the Ca2+ activation site. The Y523S mutation elicits channel preactivation in the absence of any activator and full opening at 1.5 μM free Ca2+, increasing by ∼20-fold the potency of Ca2+ to activate the channel compared with RyR1 wild type (WT). This study identified a preactivated pathological state of RyR1 and a long-range lever that may work as a molecular switch to open the channel. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. A case of dermatomyositis in a patient with central core disease: unusual association with autoimmunity and genetic muscle disease
- Author
-
Min Jung Kim, Mi Hyeon Kim, Sung-Hye Park, and Yeong Wook Song
- Subjects
Dermatomyositis ,Central core disease ,Congenital myopathy ,Pediatrics ,RJ1-570 ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Dermatomyositis is an inflammatory muscle disease caused by immune-mediated muscle injury, and central core disease (CCD) is a congenital myopathy associated with disturbed intracellular calcium homeostasis and excitation-contraction coupling. To date, CCD has not been reported to have autoantibodies or coexist with inflammatory myopathy. Case presentation Here, we described the case of a 25-year-old woman who had progressive proximal muscle weakness, myalgia, pruritic macular rash, skin ulcers, and calcinosis. Dermatomyositis was initially suspected based on the clinical symptoms accompanied by elevated muscle enzyme levels, electromyography abnormalities, and a positive antinuclear antibody test. However, the patient’s muscle biopsy revealed the characteristic findings of both dermatomyositis and CCD, suggesting that dermatomyositis occurred in this patient with previously asymptomatic CCD. The patient did not have any pathogenic gene mutations associated with congenital myopathy, including RYR1 and SEPN1 in targeted next-generation sequencing. She received high-dose glucocorticoid therapy and azathioprine with a significant improvement in muscle strength. Conclusions We present a case of rare coexistence of dermatomyositis and CCD. Clinicians should be aware that patients with CCD may have inflammatory myopathy that responds well to immunosuppressive therapy.
- Published
- 2021
- Full Text
- View/download PDF
10. Central Core Disease: Facial Weakness Differentiating Biallelic from Monoallelic Forms.
- Author
-
Cotta, Ana, Souza, Lucas Santos, Carvalho, Elmano, Feitosa, Leticia Nogueira, Cunha Jr., Antonio, Navarro, Monica Machado, Valicek, Jaquelin, Menezes, Miriam Melo, Neves, Simone Vilela Nunes, Xavier-Neto, Rafael, Vargas, Antonio Pedro, Takata, Reinaldo Issao, Paim, Julia Filardi, and Vainzof, Mariz
- Subjects
- *
GENETIC counseling , *NEUROMUSCULAR diseases , *FAMILY counseling , *NUCLEOTIDE sequencing , *MEDICAL screening , *ANDROGEN receptors , *NEMALINE myopathy - Abstract
Central Core Disease (CCD) is a genetic neuromuscular disorder characterized by the presence of cores in muscle biopsy. The inheritance has been described as predominantly autosomal dominant (AD), and the disease may present as severe neonatal or mild adult forms. Here we report clinical and molecular data on a large cohort of Brazilian CCD patients, including a retrospective clinical analysis and molecular screening for RYR1 variants using Next-Generation Sequencing (NGS). We analyzed 27 patients from 19 unrelated families: four families (11 patients) with autosomal dominant inheritance (AD), two families (3 patients) with autosomal recessive (AR), and 13 sporadic cases. Biallelic RYR1 variants were found in six families (two AR and four sporadic cases) of the 14 molecularly analyzed families (~43%), suggesting a higher frequency of AR inheritance than expected. None of these cases presented a severe phenotype. Facial weakness was more common in biallelic than in monoallelic patients (p = 0.0043) and might be a marker for AR forms. NGS is highly effective for the identification of RYR1 variants in CCD patients, allowing the discovery of a higher proportion of AR cases with biallelic mutations. These data have important implications for the genetic counseling of the families. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
11. Expanding the clinical-pathological and genetic spectrum of RYR1-related congenital myopathies with cores and minicores: an Italian population study.
- Author
-
Fusto, Aurora, Cassandrini, Denise, Fiorillo, Chiara, Codemo, Valentina, Astrea, Guja, D'Amico, Adele, Maggi, Lorenzo, Magri, Francesca, Pane, Marika, Tasca, Giorgio, Sabbatini, Daniele, Bello, Luca, Battini, Roberta, Bernasconi, Pia, Fattori, Fabiana, Bertini, Enrico Silvio, Comi, Giacomo, Messina, Sonia, Mongini, Tiziana, and Moroni, Isabella
- Subjects
- *
MUSCLE weakness , *MUSCLE diseases , *RYANODINE receptors , *STRUCTURAL bioinformatics , *SYMPTOMS , *MISSENSE mutation - Abstract
Mutations in the RYR1 gene, encoding ryanodine receptor 1 (RyR1), are a well-known cause of Central Core Disease (CCD) and Multi-minicore Disease (MmD). We screened a cohort of 153 patients carrying an histopathological diagnosis of core myopathy (cores and minicores) for RYR1 mutation. At least one RYR1 mutation was identified in 69 of them and these patients were further studied. Clinical and histopathological features were collected. Clinical phenotype was highly heterogeneous ranging from asymptomatic or paucisymptomatic hyperCKemia to severe muscle weakness and skeletal deformity with loss of ambulation. Sixty-eight RYR1 mutations, generally missense, were identified, of which 16 were novel. The combined analysis of the clinical presentation, disease progression and the structural bioinformatic analyses of RYR1 allowed to associate some phenotypes to mutations in specific domains. In addition, this study highlighted the structural bioinformatics potential in the prediction of the pathogenicity of RYR1 mutations. Further improvement in the comprehension of genotype–phenotype relationship of core myopathies can be expected in the next future: the actual lack of the human RyR1 crystal structure paired with the presence of large intrinsically disordered regions in RyR1, and the frequent presence of more than one RYR1 mutation in core myopathy patients, require designing novel investigation strategies to completely address RyR1 mutation effect. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. In vivo RyR1 reduction in muscle triggers a core-like myopathy
- Author
-
Laurent Pelletier, Anne Petiot, Julie Brocard, Benoit Giannesini, Diane Giovannini, Colline Sanchez, Lauriane Travard, Mathilde Chivet, Mathilde Beaufils, Candice Kutchukian, David Bendahan, Daniel Metzger, Clara Franzini Armstrong, Norma B. Romero, John Rendu, Vincent Jacquemond, Julien Fauré, and Isabelle Marty
- Subjects
Ryanodine receptor ,Calcium ,Skeletal muscle ,Excitation–contraction coupling ,Congenital myopathies ,Central core disease ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Mutations in the RYR1 gene, encoding the skeletal muscle calcium channel RyR1, lead to congenital myopathies, through expression of a channel with abnormal permeability and/or in reduced amount, but the direct functional whole organism consequences of exclusive reduction in RyR1 amount have never been studied. We have developed and characterized a mouse model with inducible muscle specific RYR1 deletion. Tamoxifen-induced recombination in the RYR1 gene at adult age resulted in a progressive reduction in the protein amount reaching a stable level of 50% of the initial amount, and was associated with a progressive muscle weakness and atrophy. Measurement of calcium fluxes in isolated muscle fibers demonstrated a reduction in the amplitude of RyR1-related calcium release mirroring the reduction in the protein amount. Alterations in the muscle structure were observed, with fibers atrophy, abnormal mitochondria distribution and membrane remodeling. An increase in the expression level of many proteins was observed, as well as an inhibition of the autophagy process. This model demonstrates that RyR1 reduction is sufficient to recapitulate most features of Central Core Disease, and accordingly similar alterations were observed in muscle biopsies from Dusty Core Disease patients (a subtype of Central Core Disease), pointing to common pathophysiological mechanisms related to RyR1 reduction.
- Published
- 2020
- Full Text
- View/download PDF
13. Preclinical model systems of ryanodine receptor 1-related myopathies and malignant hyperthermia: a comprehensive scoping review of works published 1990–2019
- Author
-
Tokunbor A. Lawal, Emily S. Wires, Nancy L. Terry, James J. Dowling, and Joshua J. Todd
- Subjects
Ryanodine receptor ,RYR1 ,Congenital myopathy ,Central core disease ,Preclinical ,Mouse ,Medicine - Abstract
Abstract Background Pathogenic variations in the gene encoding the skeletal muscle ryanodine receptor (RyR1) are associated with malignant hyperthermia (MH) susceptibility, a life-threatening hypermetabolic condition and RYR1-related myopathies (RYR1-RM), a spectrum of rare neuromuscular disorders. In RYR1-RM, intracellular calcium dysregulation, post-translational modifications, and decreased protein expression lead to a heterogenous clinical presentation including proximal muscle weakness, contractures, scoliosis, respiratory insufficiency, and ophthalmoplegia. Preclinical model systems of RYR1-RM and MH have been developed to better understand underlying pathomechanisms and test potential therapeutics. Methods We conducted a comprehensive scoping review of scientific literature pertaining to RYR1-RM and MH preclinical model systems in accordance with the PRISMA Scoping Reviews Checklist and the framework proposed by Arksey and O’Malley. Two major electronic databases (PubMed and EMBASE) were searched without language restriction for articles and abstracts published between January 1, 1990 and July 3, 2019. Results Our search yielded 5049 publications from which 262 were included in this review. A majority of variants tested in RYR1 preclinical models were localized to established MH/central core disease (MH/CCD) hot spots. A total of 250 unique RYR1 variations were reported in human/rodent/porcine models with 95% being missense substitutions. The most frequently reported RYR1 variant was R614C/R615C (human/porcine total n = 39), followed by Y523S/Y524S (rabbit/mouse total n = 30), I4898T/I4897T/I4895T (human/rabbit/mouse total n = 20), and R163C/R165C (human/mouse total n = 18). The dyspedic mouse was utilized by 47% of publications in the rodent category and its RyR1-null (1B5) myotubes were transfected in 23% of publications in the cellular model category. In studies of transfected HEK-293 cells, 57% of RYR1 variations affected the RyR1 channel and activation core domain. A total of 15 RYR1 mutant mouse strains were identified of which ten were heterozygous, three were compound heterozygous, and a further two were knockout. Porcine, avian, zebrafish, C. elegans, canine, equine, and drosophila model systems were also reported. Conclusions Over the past 30 years, there were 262 publications on MH and RYR1-RM preclinical model systems featuring more than 200 unique RYR1 variations tested in a broad range of species. Findings from these studies have set the foundation for therapeutic development for MH and RYR1-RM.
- Published
- 2020
- Full Text
- View/download PDF
14. A review of core myopathy: central core disease, multiminicore disease, dusty core disease, and core-rod myopathy.
- Author
-
Ogasawara, Masashi and Nishino, Ichizo
- Subjects
- *
NEMALINE myopathy , *MUSCLE diseases , *DIAGNOSIS - Abstract
• Pathological review of central core disease and dusty core disease. • Up-to-date genetical and pathological review of multiminicore disease and core-rod myopathy. • Charasteristic pictures of genetically confirmed core myopahites. Core myopathies are clinically, pathologically, and genetically heterogeneous muscle diseases. Their onset and clinical severity are variable. Core myopathies are diagnosed by muscle biopsy showing focally reduced oxidative enzyme activity and can be pathologically divided into central core disease, multiminicore disease, dusty core disease, and core-rod myopathy. Although RYR1- related myopathy is the most common core myopathy, an increasing number of other causative genes have been reported, including SELENON, MYH2, MYH7, TTN, CCDC78, UNC45B, ACTN2, MEGF10, CFL2, KBTBD13, and TRIP4. Furthermore, the genes originally reported to cause nemaline myopathy, namely ACTA1, NEB , and TNNT1 , have been recently associated with core-rod myopathy. Genetic analysis allows us to diagnose each core myopathy more accurately. In this review, we aim to provide up-to-date information about core myopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
15. A case of dermatomyositis in a patient with central core disease: unusual association with autoimmunity and genetic muscle disease.
- Author
-
Kim, Min Jung, Kim, Mi Hyeon, Park, Sung-Hye, and Song, Yeong Wook
- Subjects
- *
DERMATOMYOSITIS , *MUSCLE diseases , *GENETIC disorders , *MUSCLE weakness , *MEDICAL personnel , *SYMPTOMS - Abstract
Background: Dermatomyositis is an inflammatory muscle disease caused by immune-mediated muscle injury, and central core disease (CCD) is a congenital myopathy associated with disturbed intracellular calcium homeostasis and excitation-contraction coupling. To date, CCD has not been reported to have autoantibodies or coexist with inflammatory myopathy. Case presentation: Here, we described the case of a 25-year-old woman who had progressive proximal muscle weakness, myalgia, pruritic macular rash, skin ulcers, and calcinosis. Dermatomyositis was initially suspected based on the clinical symptoms accompanied by elevated muscle enzyme levels, electromyography abnormalities, and a positive antinuclear antibody test. However, the patient's muscle biopsy revealed the characteristic findings of both dermatomyositis and CCD, suggesting that dermatomyositis occurred in this patient with previously asymptomatic CCD. The patient did not have any pathogenic gene mutations associated with congenital myopathy, including RYR1 and SEPN1 in targeted next-generation sequencing. She received high-dose glucocorticoid therapy and azathioprine with a significant improvement in muscle strength. Conclusions: We present a case of rare coexistence of dermatomyositis and CCD. Clinicians should be aware that patients with CCD may have inflammatory myopathy that responds well to immunosuppressive therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
16. Perioperative management of bronchoscopy in a child patient with central core disease: A case report and literature review.
- Author
-
Li, Hong-bo, Ji, Xiang-Lin, Nie, Pei-he, Sun, Neng-hong, and Li, Rui-hua
- Abstract
We described the perioperative management of a child patient with central core disease for bronchoscopy with bronchoalveolar lavage. It is safe to avoid triggering agents (volatile anesthetics and succinylcholine) probably in preventing this appearance of malignant hyperthermia (MH). It is important to recognize potential complications and know how to prevent and manage them in patients with this condition. A 5-year-old boy (weight: 8.8 kg; height: 63 cm) presented to the pediatric department after five days of intermittent fever (highest body temperature is 39.3 °C) and cough, and aggravation 1 day, meanwhile he had phlegm in throat but he couldn't cough out. The child was found to have motor retardation at his one-month-old physical examination, then genetic analysis showed central core disease. Bronchoscopy with bronchoalveolar lavage was performed for better treatment under the premise of symptomatic treatment. The patients with central core disease are particularly to develop malignant hyperthermia, so adequate precautions are in place to prevent and treat MH before anesthetic induction. The anesthesiologists need to make adequate preoperative anesthesia management strategies to ensure the safety of the child with central core disease for bronchoscopy with bronchoalveolar lavage. The child was discharged from the hospital one week after anti-inflammatory and anti-asthmatic treatment. We summarized the anesthetic precautions and management in patients with central core disease, meanwhile we offered some suggestions about anesthetic focus on bronchoscopy with bronchoalveolar lavage. • Good perioperative management of bronchoscopy in a child patient with central core disease is necessary for good prognosis. • We summarized the anesthetic precautions and management in patients with central core disease. • We offered some suggestions about anesthetic focus on bronchoscopy with bronchoalveolar lavage. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. ‘Dusty core disease’ (DuCD): expanding morphological spectrum of RYR1 recessive myopathies
- Author
-
Matteo Garibaldi, John Rendu, Julie Brocard, Emmanuelle Lacene, Julien Fauré, Guy Brochier, Maud Beuvin, Clemence Labasse, Angeline Madelaine, Edoardo Malfatti, Jorge Alfredo Bevilacqua, Fabiana Lubieniecki, Soledad Monges, Ana Lia Taratuto, Jocelyn Laporte, Isabelle Marty, Giovanni Antonini, and Norma Beatriz Romero
- Subjects
RYR1 recessive ,Dusty Core Disease ,Central Core Disease ,Congenital Myopathy ,Centronuclear myopathy ,Ryanodine receptor ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Several morphological phenotypes have been associated to RYR1-recessive myopathies. We recharacterized the RYR1-recessive morphological spectrum by a large monocentric study performed on 54 muscle biopsies from a large cohort of 48 genetically confirmed patients, using histoenzymology, immunohistochemistry, and ultrastructural studies. We also analysed the level of RyR1 expression in patients’ muscle biopsies. We defined “dusty cores” the irregular areas of myofibrillar disorganisation characterised by a reddish-purple granular material deposition with uneven oxidative stain and devoid of ATPase activity, which represent the characteristic lesion in muscle biopsy in 54% of patients. We named Dusty Core Disease (DuCD) the corresponding entity of congenital myopathy. Dusty cores had peculiar histological and ultrastructural characteristics compared to the other core diseases. DuCD muscle biopsies also showed nuclear centralization and type1 fibre predominance. Dusty cores were not observed in other core myopathies and centronuclear myopathies. The other morphological groups in our cohort of patients were: Central Core (CCD: 21%), Core-Rod (C&R:15%) and Type1 predominance “plus” (T1P+:10%). DuCD group was associated to an earlier disease onset, a more severe clinical phenotype and a lowest level of RyR1 expression in muscle, compared to the other groups. Variants located in the bridge solenoid and the pore domains were more frequent in DuCD patients. In conclusion, DuCD is the most frequent histopathological presentation of RYR1-recessive myopathies. Dusty cores represent the unifying morphological lesion among the DuCD pathology spectrum and are the morphological hallmark for the recessive form of disease.
- Published
- 2019
- Full Text
- View/download PDF
18. Neuromuscular Disease
- Author
-
Cravero, Joseph P., Holzman, Robert S., editor, Mancuso, Thomas J., editor, Cravero, Joseph P., editor, and DiNardo, James A., editor
- Published
- 2017
- Full Text
- View/download PDF
19. Myopathies: Congenital, Metabolic and Mitochondrial, and Channelopathies
- Author
-
Crochetiere, Chantal, Lidzborski, Elsa, and Mankowitz, Suzanne K. W., editor
- Published
- 2018
- Full Text
- View/download PDF
20. Novel ACTA1 mutation causes late-presenting nemaline myopathy with unusual dark cores.
- Author
-
Garibaldi, Matteo, Fattori, Fabiana, Pennisi, Elena Maria, Merlonghi, Gioia, Fionda, Laura, Vanoli, Fiammetta, Leonardi, Luca, Bucci, Elisabetta, Morino, Stefania, Micaloni, Andrea, Tartaglione, Tommaso, Uijterwijk, Bas, Zierikzee, Martijn, Ottenheijm, Coen, Bertini, Enrico Silvio, Stoppacciaro, Antonella, Raffa, Salvatore, Salvetti, Marco, and Antonini, Giovanni
- Subjects
- *
NEMALINE myopathy , *MUSCLE weakness , *RECTUS abdominis muscles , *SKELETAL muscle , *ELECTRON microscopy , *GENES , *PLANTARFLEXION - Abstract
• A novel dominant mutation (Gly50Ser) in ACTA1 causes mild late onset core-rod myopathy. • Peculiar areas of fuzzy-dark material deposition corresponded to unstructured cores. • Affected members showed mild weakness with high arched palate and sternocleidomastoideus atrophy. • Muscle MRI showed the same pattern of fibro-fatty replacement in all examined probands. • Fiber contractility essay demonstrates a higher calcium sensitivity of force in type i fibers. ACTA1 gene encodes the skeletal muscle alpha-actin, the core of thin filaments of the sarcomere. ACTA1 mutations are responsible of several muscle disorders including nemaline, cores, actin aggregate myopathies and fiber-type disproportion. We report clinical, muscle imaging, histopatological and genetic data of an Italian family carrying a novel ACTA1 mutation. All affected members showed a late-presenting, diffuse muscle weakness with sternocleidomastoideus and temporalis atrophy. Mild dysmorphic features were also detected. The most affected muscles by muscle MRI were rectus abdominis, gluteus minimus, vastus intermedius and both gastrocnemii. Muscle biopsy showed the presence of nemaline bodies with several unusual dark areas at Gomori Trichrome, corresponding to unstructured cores with abundant electrodense material by electron microscopy. The molecular analysis revealed missense variant c.148G> A ; p.(Gly50Ser) in the exon 3 of ACTA1 , segregating with affected members in the family. We performed a functional essay of fibre contractility showing a higher pCa 50 (a measure of the calcium sensitivity of force) of type 1 fibers compared to control subjects' type 1 muscle fibers. Our findings expand the clinico-pathological spectrum of ACTA1 -related congenital myopathies and the genetic spectrum of core-rod myopathies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
21. Intracellular calcium leak as a therapeutic target for RYR1-related myopathies.
- Author
-
Kushnir, Alexander, Todd, Joshua J., Witherspoon, Jessica W., Yuan, Qi, Reiken, Steven, Lin, Harvey, Munce, Ross H., Wajsberg, Benjamin, Melville, Zephan, Clarke, Oliver B., Wedderburn-Pugh, Kaylee, Wronska, Anetta, Razaqyar, Muslima S., Chrismer, Irene C., Shelton, Monique O., Mankodi, Ami, Grunseich, Christopher, Tarnopolsky, Mark A., Tanji, Kurenai, and Hirano, Michio
- Subjects
- *
RYANODINE receptors , *INTRACELLULAR calcium , *NEUROMUSCULAR diseases , *MUSCLE diseases , *SARCOPLASMIC reticulum , *SKELETAL muscle - Abstract
RYR1 encodes the type 1 ryanodine receptor, an intracellular calcium release channel (RyR1) on the skeletal muscle sarcoplasmic reticulum (SR). Pathogenic RYR1 variations can destabilize RyR1 leading to calcium leak causing oxidative overload and myopathy. However, the effect of RyR1 leak has not been established in individuals with RYR1-related myopathies (RYR1-RM), a broad spectrum of rare neuromuscular disorders. We sought to determine whether RYR1-RM affected individuals exhibit pathologic, leaky RyR1 and whether variant location in the channel structure can predict pathogenicity. Skeletal muscle biopsies were obtained from 17 individuals with RYR1-RM. Mutant RyR1 from these individuals exhibited pathologic SR calcium leak and increased activity of calcium-activated proteases. The increased calcium leak and protease activity were normalized by ex-vivo treatment with S107, a RyR stabilizing Rycal molecule. Using the cryo-EM structure of RyR1 and a new dataset of > 2200 suspected RYR1-RM affected individuals we developed a method for assigning pathogenicity probabilities to RYR1 variants based on 3D co-localization of known pathogenic variants. This study provides the rationale for a clinical trial testing Rycals in RYR1-RM affected individuals and introduces a predictive tool for investigating the pathogenicity of RYR1 variants of uncertain significance. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. Preclinical model systems of ryanodine receptor 1-related myopathies and malignant hyperthermia: a comprehensive scoping review of works published 1990-2019.
- Author
-
Lawal, Tokunbor A., Wires, Emily S., Terry, Nancy L., Dowling, James J., and Todd, Joshua J.
- Subjects
- *
MALIGNANT hyperthermia , *ANIMAL models in research , *RYANODINE receptors , *SCIENTIFIC literature , *NEUROMUSCULAR diseases , *MUSCLE diseases - Abstract
Background: Pathogenic variations in the gene encoding the skeletal muscle ryanodine receptor (RyR1) are associated with malignant hyperthermia (MH) susceptibility, a life-threatening hypermetabolic condition and RYR1-related myopathies (RYR1-RM), a spectrum of rare neuromuscular disorders. In RYR1-RM, intracellular calcium dysregulation, post-translational modifications, and decreased protein expression lead to a heterogenous clinical presentation including proximal muscle weakness, contractures, scoliosis, respiratory insufficiency, and ophthalmoplegia. Preclinical model systems of RYR1-RM and MH have been developed to better understand underlying pathomechanisms and test potential therapeutics.Methods: We conducted a comprehensive scoping review of scientific literature pertaining to RYR1-RM and MH preclinical model systems in accordance with the PRISMA Scoping Reviews Checklist and the framework proposed by Arksey and O'Malley. Two major electronic databases (PubMed and EMBASE) were searched without language restriction for articles and abstracts published between January 1, 1990 and July 3, 2019.Results: Our search yielded 5049 publications from which 262 were included in this review. A majority of variants tested in RYR1 preclinical models were localized to established MH/central core disease (MH/CCD) hot spots. A total of 250 unique RYR1 variations were reported in human/rodent/porcine models with 95% being missense substitutions. The most frequently reported RYR1 variant was R614C/R615C (human/porcine total n = 39), followed by Y523S/Y524S (rabbit/mouse total n = 30), I4898T/I4897T/I4895T (human/rabbit/mouse total n = 20), and R163C/R165C (human/mouse total n = 18). The dyspedic mouse was utilized by 47% of publications in the rodent category and its RyR1-null (1B5) myotubes were transfected in 23% of publications in the cellular model category. In studies of transfected HEK-293 cells, 57% of RYR1 variations affected the RyR1 channel and activation core domain. A total of 15 RYR1 mutant mouse strains were identified of which ten were heterozygous, three were compound heterozygous, and a further two were knockout. Porcine, avian, zebrafish, C. elegans, canine, equine, and drosophila model systems were also reported.Conclusions: Over the past 30 years, there were 262 publications on MH and RYR1-RM preclinical model systems featuring more than 200 unique RYR1 variations tested in a broad range of species. Findings from these studies have set the foundation for therapeutic development for MH and RYR1-RM. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
23. Single Amino Acid Changes in the Ryanodine Receptor in the Human Population Have Effects In Vivo on Caenorhabditis elegans Neuro-Muscular Function.
- Author
-
Graham, Brittany, Shaw, Marie-Anne, and Hope, Ian A.
- Subjects
RYANODINE receptors ,CAENORHABDITIS elegans ,SUGAMMADEX ,POPULATION ,CALCIUM channels ,AMINO acid residues ,ION channels - Abstract
The ryanodine receptor mediates intracellular calcium ion release with excitation of nerve and muscle cells. Ryanodine receptor missense variants cause a number of myopathologies, such as malignant hyperthermia, and have been linked with various neuropathologies, including Alzheimer's disease. We characterized the consequences of ryanodine receptor variants in vivo. Eight Caenorhabditis elegans strains, with ryanodine receptor modifications equivalent to human myopathic RYR1 variants, were generated by genome editing. In humans, these variants are rare and confer sensitivity to the inhalational anaesthetic halothane when heterozygous. Increased sensitivity to halothane was found in both homozygous and heterozygous C. elegans. Close analysis revealed distinct subtle locomotion defects, due to the different single amino acid residue changes, even in the absence of the external triggering agent. Distinct pre- and postsynaptic consequences of the variants were characterized through the responses to cholinergic pharmacological agents. The range of phenotypes reflects the complexity of the regulatory inputs to the ryanodine receptor and the criticality of the calcium ion channel opening properties, in different cell types and with age. Ryanodine receptors with these single amino acid residue changes still function as calcium ion channels, but with altered properties which are likely to have subtle consequences for human carriers of such variants. The long-term consequences of subtly altered calcium ion signalling could be cumulative and may be focussed in the smaller nerve cells rather than the more robust muscle cells. It was important to assess phenotypes in vivo to properly appreciate consequences for a whole organism. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
24. Hereditary Neuromuscular Diseases and Cardiac Involvement
- Author
-
van der Kooi, A. J., Wahbi, K., Bonne, G., de Visser, M., Baars, Hubert F., editor, Doevendans, Pieter A. F. M., editor, Houweling, Arjan C., editor, and van Tintelen, J. Peter, editor
- Published
- 2016
- Full Text
- View/download PDF
25. Rhabdomyosarcoma Associated with Core Myopathy/Malignant Hyperthermia: Combined Effect of Germline Variants in RYR1 and ASPSCR1 May Play a Role
- Author
-
Silva, Pamela V. Andrade, Joilson M. Santos, Anne C. B. Teixeira, Vanessa F. Sogari, Michelle S. Almeida, Fabiano M. Callegari, Ana C. V. Krepischi, Acary S. B. Oliveira, Mariz Vainzof, and Helga C. A.
- Subjects
malignant hyperthermia ,rhabdomyosarcoma ,central core disease ,ryanodine receptor ,ASPSCR1 protein - Abstract
Rhabdomyosarcomas have been described in association with thyroid disease, dermatomyositis, Duchenne muscular dystrophy, and in muscular dystrophy models but not in patients with ryanodine receptor-1 gene (RYR1) pathogenic variants. We described here an 18-year-old male who reported a cervical nodule. Magnetic resonance images revealed a mass in the ethmoidal sinus corresponding to rhabdomyosarcoma. As his father died from malignant hyperthermia (MH), an in vitro contracture test was conducted and was positive for MH susceptibility. Muscle histopathological analysis in the biopsy showed the presence of cores. Molecular analysis using NGS sequencing identified germline variants in the RYR1 and ASPSCR1 (alveolar soft part sarcoma) genes. This report expands the spectrum of diseases associated with rhabdomyosarcomas and a possible differential diagnosis of soft tissue tumors in patients with RYR1 variants.
- Published
- 2023
- Full Text
- View/download PDF
26. Rycal S48168 (ARM210) for RYR1 -related myopathies: a phase one, open-label, dose-escalation trial.
- Author
-
Todd JJ, Lawal TA, Chrismer IC, Kokkinis A, Grunseich C, Jain MS, Waite MR, Biancavilla V, Pocock S, Brooks K, Mendoza CJ, Norato G, Cheung K, Riekhof W, Varma P, Colina-Prisco C, Emile-Backer M, Meilleur KG, Marks AR, Webb Y, Marcantonio EE, Foley AR, Bönnemann CG, and Mohassel P
- Abstract
Background: RYR1 -related myopathies ( RYR1 -RM) are caused by pathogenic variants in the RYR1 gene which encodes the type 1 ryanodine receptor (RyR1). RyR1 is the sarcoplasmic reticulum (SR) calcium release channel that mediates excitation-contraction coupling in skeletal muscle. RyR1 sub-conductance, SR calcium leak, reduced RyR1 expression, and oxidative stress often contribute to RYR1 -RM pathogenesis. Loss of RyR1-calstabin1 association, SR calcium leak, and increased RyR1 open probability were observed in 17 RYR1 -RM patient skeletal muscle biopsies and improved following ex vivo treatment with Rycal compounds. Thus, we initiated a first-in-patient trial of Rycal S48168 (ARM210) in ambulatory adults with genetically confirmed RYR1 -RM., Methods: Participants received 120 mg (n = 3) or 200 mg (n = 4) S48168 (ARM210) daily for 29 days. The primary endpoint was safety and tolerability. Exploratory endpoints included S48168 (ARM210) pharmacokinetics (PK), target engagement, motor function measure (MFM)-32, hand grip and pinch strength, timed functional tests, PROMIS fatigue scale, semi-quantitative physical exam strength measurements, and oxidative stress biomarkers. The trial was registered with clinicaltrials.gov (NCT04141670) and was conducted at the National Institutes of Health Clinical Center between October 28, 2019 and December 12, 2021., Findings: S48168 (ARM210) was well-tolerated, did not cause any serious adverse events, and exhibited a dose-dependent PK profile. Three of four participants who received the 200 mg/day dose reported improvements in PROMIS-fatigue at 28 days post-dosing, and also demonstrated improved proximal muscle strength on physical examination., Interpretation: S48168 (ARM210) demonstrated favorable safety, tolerability, and PK, in RYR1 -RM affected individuals. Most participants who received 200 mg/day S48168 (ARM210) reported decreased fatigue, a key symptom of RYR1 -RM. These results set the foundation for a randomized, double-blind, placebo-controlled proof of concept trial to determine efficacy of S48168 (ARM210) in RYR1 -RM., Funding: NINDS and NINR Intramural Research Programs, NIH Clinical Center Bench to Bedside Award (2017-551673), ARMGO Pharma Inc., and its development partner Les Laboratoires Servier., Competing Interests: E.E.M is an employee of ARMGO Pharma, Inc., during the conduct of the study; an employee of ARMGO Pharma, Inc., outside the submitted work; In addition, E.E.M has a patent US2023/0372358 pending. Y.W is an employee of ARMGO Pharma, Inc. In addition, Y.W has a patent US8853198 issued. A.R.M. is a co-founder of ARMGO Pharma Inc, chairs the Scientific Advisory Board of ARMGO Pharma Inc., and holds stock in the company. All other authors declare no conflict of interest with the research in this manuscript., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
27. Anesthetic Management of a Patient With Central Core Disease Undergoing Thoracoscopic Lung Resection: The Importance of Neuromuscular Monitoring at the Masseter Muscle.
- Author
-
Baba H, Wakabayashi R, Ichiyanagi H, Suzuki A, and Sato N
- Abstract
Central core disease is a rare muscular disorder in which anesthetic considerations for the prevention of malignant hyperthermia and for avoidance of residual neuromuscular block are required. A 63-year-old woman with central core disease underwent thoracoscopic sublobar lung resection under total IV anesthesia with a prepared anesthetic workstation. The rocuronium-induced neuromuscular block was monitored by using acceleromyography at the left adductor pollicis muscle and the right masseter muscle. The recovery of neuromuscular block at the masseter was slower than that at the adductor pollicis. The patient showed no symptoms of malignant hyperthermia and residual neuromuscular block and had an uneventful postoperative course. In the present case, malignant hyperthermia was successfully prevented with general anesthesia that is free of triggering agents using a prepared anesthetic machine. The authors speculate that the masseter may be an auxiliary site for neuromuscular monitoring to ensure recovery from neuromuscular block in patients with central core disease., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2024, Baba et al.)
- Published
- 2024
- Full Text
- View/download PDF
28. Ryanodine Receptor 1 and Associated Pathologies
- Author
-
Fauré, Julien, Lunardi, Joël, Monnier, Nicole, Marty, Isabelle, Weiss, Norbert, editor, and Koschak, Alexandra, editor
- Published
- 2014
- Full Text
- View/download PDF
29. Malignant Hyperthermia
- Author
-
Bertorini, Tulio E., Katirji, Bashar, editor, Kaminski, Henry J., editor, and Ruff, Robert L., editor
- Published
- 2014
- Full Text
- View/download PDF
30. A central core disease mutation in the Ca2+-binding site of skeletal muscle ryanodine receptor impairs single-channel regulation.
- Author
-
Chirasani, Venkat R., Le Xu, Addis, Hannah G., Pasek, Daniel A., Dokholyan, Nikolay V., Meissner, Gerhard, and Naohiro Yamaguchi
- Abstract
Cryoelectron microscopy and mutational analyses have shown that type 1 ryanodine receptor (RyR1) amino acid residues RyR1-E3893, -E3967, and -T5001 are critical for Ca2+-mediated activation of skeletal muscle Ca2+ release channel. De novo missense mutation RyR1-Q3970K in the secondary binding sphere of Ca2+ was reported in association with central core disease (CCD) in a 2-yr-old boy. Here, we characterized recombinant RyR1-Q3970K mutant by cellular Ca2+ release measurements, single-channel recordings, and computational methods. Caffeine-induced Ca2+ release studies indicated that RyR1-Q3970K formed caffeine-sensitive, Ca2+-conducting channel in HEK293 cells. However, in single-channel recordings, RyR1-Q3970K displayed low Ca2+-dependent channel activity and greatly reduced activation by caffeine or ATP. A RyR1-Q3970E mutant corresponds to missense mutation RyR2-Q3925E associated with arrhythmogenic syndrome in cardiac muscle. RyR1-Q3970E also formed caffeine-induced Ca2+ release in HEK293 cells and exhibited low activity in the presence of the activating ligand Ca2+ but, in contrast to RyR1-Q3970K, was activated by ATP and caffeine in single-channel recordings. Computational analyses suggested distinct structural rearrangements in the secondary binding sphere of Ca2+ of the two mutants, whereas the interaction of Ca2+ with directly interacting RyR1 amino acid residues Glu3893, Glu3967, and Thr5001 was only minimally affected. We conclude that RyR1-Q3970 has a critical role in Ca2+-dependent activation of RyR1 and that a missense RyR1-Q3970K mutant may give rise to myopathy in skeletal muscle. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
31. Congenital Myopathies
- Author
-
Quijano-Roy, Susana, Avila-Smirnow, Daniela, Carlier, Robert-Yves, Bevilacqua, Jorge A., Romero, Norma Beatriz, Fischer, Dirk, Wattjes, Mike P., editor, and Fischer, Dirk, editor
- Published
- 2013
- Full Text
- View/download PDF
32. Ryanodine Receptor Physiology and Its Role in Disease
- Author
-
Lanner, Johanna T. and Islam, Md. Shahidul, editor
- Published
- 2012
- Full Text
- View/download PDF
33. Hereditary Neuromuscular Diseases and Cardiac Involvement
- Author
-
van der Kooi, A. J., de Visser, M., Baars, H.F., editor, Doevendans, P.A.F.M., editor, and van der Smagt, J.J., editor
- Published
- 2011
- Full Text
- View/download PDF
34. Calcium Channelopathies: Structural Insights into Disorders of the Muscle Excitation–Contraction Complex.
- Author
-
Pancaroglu, Raika and Van Petegem, Filip
- Abstract
Ion channels are membrane proteins responsible for the passage of ions down their electrochemical gradients and across biological membranes. In this, they generate and shape action potentials and provide secondary messengers for various signaling pathways. They are often part of larger complexes containing auxiliary subunits and regulatory proteins. Channelopathies arise from mutations in the genes encoding ion channels or their associated proteins. Recent advances in cryo-electron microscopy have resulted in an explosion of ion channel structures in multiple states, generating a wealth of new information on channelopathies. Disease-associated mutations fall into different categories, interfering with ion permeation, protein folding, voltage sensing, ligand and protein binding, and allosteric modulation of channel gating. Prime examples of these are Ca2+-selective channels expressed in myocytes, for which multiple structures in distinct conformational states have recently been uncovered. We discuss the latest insights into these calcium channelopathies from a structural viewpoint. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
35. Ryanodine Receptor 1-Related Myopathies: Diagnostic and Therapeutic Approaches.
- Author
-
Lawal, Tokunbor A., Todd, Joshua J., and Meilleur, Katherine G.
- Subjects
CALCIUM metabolism ,MUSCLE disease treatment ,DIAGNOSIS of muscle diseases ,ANIMALS ,CALCIUM ,MUSCLE diseases - Abstract
Ryanodine receptor type 1-related myopathies (RYR1-RM) are the most common class of congenital myopathies. Historically, RYR1-RM classification and diagnosis have been guided by histopathologic findings on muscle biopsy. Main histological subtypes of RYR1-RM include central core disease, multiminicore disease, core-rod myopathy, centronuclear myopathy, and congenital fiber-type disproportion. A range of RYR1-RM clinical phenotypes has also emerged more recently and includes King Denborough syndrome, RYR1 rhabdomyolysis-myalgia syndrome, atypical periodic paralysis, congenital neuromuscular disease with uniform type 1 fibers, and late-onset axial myopathy. This expansion of the RYR1-RM disease spectrum is due, in part, to implementation of next-generation sequencing methods, which include the entire RYR1 coding sequence rather than being restricted to hotspot regions. These methods enhance diagnostic capabilities, especially given historic limitations of histopathologic and clinical overlap across RYR1-RM. Both dominant and recessive modes of inheritance have been documented, with the latter typically associated with a more severe clinical phenotype. As with all congenital myopathies, no FDA-approved treatments exist to date. Here, we review histopathologic, clinical, imaging, and genetic diagnostic features of the main RYR1-RM subtypes. We also discuss the current state of treatments and focus on disease-modulating (nongenetic) therapeutic strategies under development for RYR1-RM. Finally, perspectives for future approaches to treatment development are broached. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
36. Expanding the clinical-pathological and genetic spectrum of RYR1-related congenital myopathies with cores and minicores: an Italian population study
- Author
-
Fusto, A., Cassandrini, D., Fiorillo, C., Codemo, V., Astrea, G., D'Amico, A., Maggi, L., Magri, F., Pane, M., Tasca, G., Sabbatini, D., Bello, L., Battini, R., Bernasconi, P., Fattori, F., Bertini, E. S., Comi, G., Messina, S., Mongini, T., Moroni, I., Panicucci, C., Berardinelli, A., Donati, A., Nigro, V., Pini, A., Giannotta, M., Dosi, C., Ricci, E., Mercuri, E., Minervini, G., Tosatto, S., Santorelli, F., Bruno, C., Pegoraro, E., Fiorillo C. (ORCID:0000-0001-7681-3567), D'Amico A., Pane M. (ORCID:0000-0002-4851-6124), Tasca G., Battini R., Fattori F., Bertini E. S., Ricci E. (ORCID:0000-0003-3092-3597), Mercuri E. (ORCID:0000-0002-9851-5365), Fusto, A., Cassandrini, D., Fiorillo, C., Codemo, V., Astrea, G., D'Amico, A., Maggi, L., Magri, F., Pane, M., Tasca, G., Sabbatini, D., Bello, L., Battini, R., Bernasconi, P., Fattori, F., Bertini, E. S., Comi, G., Messina, S., Mongini, T., Moroni, I., Panicucci, C., Berardinelli, A., Donati, A., Nigro, V., Pini, A., Giannotta, M., Dosi, C., Ricci, E., Mercuri, E., Minervini, G., Tosatto, S., Santorelli, F., Bruno, C., Pegoraro, E., Fiorillo C. (ORCID:0000-0001-7681-3567), D'Amico A., Pane M. (ORCID:0000-0002-4851-6124), Tasca G., Battini R., Fattori F., Bertini E. S., Ricci E. (ORCID:0000-0003-3092-3597), and Mercuri E. (ORCID:0000-0002-9851-5365)
- Abstract
Mutations in the RYR1 gene, encoding ryanodine receptor 1 (RyR1), are a well-known cause of Central Core Disease (CCD) and Multi-minicore Disease (MmD). We screened a cohort of 153 patients carrying an histopathological diagnosis of core myopathy (cores and minicores) for RYR1 mutation. At least one RYR1 mutation was identified in 69 of them and these patients were further studied. Clinical and histopathological features were collected. Clinical phenotype was highly heterogeneous ranging from asymptomatic or paucisymptomatic hyperCKemia to severe muscle weakness and skeletal deformity with loss of ambulation. Sixty-eight RYR1 mutations, generally missense, were identified, of which 16 were novel. The combined analysis of the clinical presentation, disease progression and the structural bioinformatic analyses of RYR1 allowed to associate some phenotypes to mutations in specific domains. In addition, this study highlighted the structural bioinformatics potential in the prediction of the pathogenicity of RYR1 mutations. Further improvement in the comprehension of genotype–phenotype relationship of core myopathies can be expected in the next future: the actual lack of the human RyR1 crystal structure paired with the presence of large intrinsically disordered regions in RyR1, and the frequent presence of more than one RYR1 mutation in core myopathy patients, require designing novel investigation strategies to completely address RyR1 mutation effect.
- Published
- 2022
37. Diseases associated with altered ryanodine receptor activity
- Author
-
DURHAM, W.J., WEHRENS, X.H.T., SOOD, S., HAMILTON, S.L., Harris, J. Robin, editor, Biswas, B.B., editor, Quinn, P., editor, Carafoli, Ernesto, editor, and Brini, Marisa, editor
- Published
- 2007
- Full Text
- View/download PDF
38. Evolution of the Ryanodine Receptor Gene Family
- Author
-
Kushnir, Alexander, Mollah, A. K. M. M., Wehrens, Xander H. T., Wehrens, Xander H. T., editor, and Marks, Andrew R., editor
- Published
- 2005
- Full Text
- View/download PDF
39. Modulation of Calcium Homeostasis by the Endoplasmic Reticulum in Health and Disease
- Author
-
Szabadkai, György, Chami, Mounia, Pinton, Paolo, Rizzuto, Rosario, Eggleton, Paul, editor, and Michalak, Marek, editor
- Published
- 2003
- Full Text
- View/download PDF
40. Familial Hypertrophic Cardiomyopathic Myosin Mutations That Affect the Actin-Myosin Interaction
- Author
-
Roopnarine, Osha, Hennig, W., editor, Thomas, David D., editor, and Dos Remedios, Cristobal G., editor
- Published
- 2002
- Full Text
- View/download PDF
41. Novel ACTA1 mutation causes late-presenting nemaline myopathy with unusual dark cores
- Author
-
Enrico Bertini, Elisabetta Bucci, Giovanni Antonini, Luca Leonardi, Marco Salvetti, Bas Uijterwijk, Antonella Stoppacciaro, Martijn Zierikzee, Salvatore Raffa, Fiammetta Vanoli, Tommaso Tartaglione, Laura Fionda, Elena Maria Pennisi, Gioia Merlonghi, Fabiana Fattori, Coen A.C. Ottenheijm, Matteo Garibaldi, Stefania Morino, Andrea Micaloni, Physiology, and ACS - Pulmonary hypertension & thrombosis
- Subjects
Adult ,Male ,Sarcomeres ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Mutation, Missense ,Central core ,Acta1 ,Muscle disorder ,Biology ,Myopathies, Nemaline ,Sarcomere ,03 medical and health sciences ,0302 clinical medicine ,Nemaline myopathy ,medicine ,Central core disease ,Humans ,Muscle, Skeletal ,Nemaline bodies ,Genetics (clinical) ,Aged ,Congenital myopathy ,Muscle Weakness ,Muscle biopsy ,medicine.diagnostic_test ,Skeletal muscle ,Muscle weakness ,Middle Aged ,medicine.disease ,Actins ,Pedigree ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Core-rod myopathy ,Mutation ,Pediatrics, Perinatology and Child Health ,Female ,Neurology (clinical) ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
ACTA1 gene encodes the skeletal muscle alpha-actin, the core of thin filaments of the sarcomere. ACTA1 mutations are responsible of several muscle disorders including nemaline, cores, actin aggregate myopathies and fiber-type disproportion. We report clinical, muscle imaging, histopatological and genetic data of an Italian family carrying a novel ACTA1 mutation. All affected members showed a late-presenting, diffuse muscle weakness with sternocleidomastoideus and temporalis atrophy. Mild dysmorphic features were also detected. The most affected muscles by muscle MRI were rectus abdominis, gluteus minimus, vastus intermedius and both gastrocnemii. Muscle biopsy showed the presence of nemaline bodies with several unusual dark areas at Gomori Trichrome, corresponding to unstructured cores with abundant electrodense material by electron microscopy. The molecular analysis revealed missense variant c.148G>A; p.(Gly50Ser) in the exon 3 of ACTA1, segregating with affected members in the family. We performed a functional essay of fibre contractility showing a higher pCa50 (a measure of the calcium sensitivity of force) of type 1 fibers compared to control subjects’ type 1 muscle fibers. Our findings expand the clinico-pathological spectrum of ACTA1-related congenital myopathies and the genetic spectrum of core-rod myopathies.
- Published
- 2021
- Full Text
- View/download PDF
42. Dominant or recessive mutations in the RYR1 gene causing central core myopathy in Brazilian patients
- Author
-
Galleni Leão, Leonardo, Santos Souza, Lucas, Nogueira, Letícia, Pavanello, Rita de Cássia Mingroni, Gurgel-Giannetti, Juliana, Reed, Umbertina C, Oliveira, Acary S.B., Cuperman, Thais, Cotta, Ana, FPaim, Julia, Zatz, Mayana, and Vainzof, Mariz
- Subjects
Adult ,Male ,Heterozygote ,Homozygote ,Inheritance Patterns ,Next Generation Sequencing ,High-Throughput Nucleotide Sequencing ,Ryanodine Receptor Calcium Release Channel ,Child, Preschool ,Mutation ,RYR1 ,Humans ,Original Article ,Female ,Myopathy, Central Core ,Child ,central core disease ,Brazil - Abstract
Central Core Disease (CCD) is an inherited neuromuscular disorder characterized by the presence of cores in muscle biopsy. CCD is caused by mutations in the RYR1 gene. This gene encodes the ryanodine receptor 1, which is an intracellular calcium release channel from the sarcoplasmic reticulum to the cytosol in response to depolarization of the plasma membrane. Mutations in this gene are also associated with susceptibility to Malignant Hyperthermia (MHS). In this study, we evaluated 20 families with clinical and histological characteristics of CCD to identify primary mutations in patients, for diagnosis and genetic counseling of the families. We identified variants in the RYR1 gene in 19/20 families. The molecular pathogenicity was confirmed in 16 of them. Most of these variants (22/23) are missense and unique in the families. Two variants were recurrent in two different families. We identified six families with biallelic mutations, five compound heterozygotes with no consanguinity, and one homozygous, with consanguineous parents, resulting in 30% of cases with possible autosomal recessive inheritance. We identified seven novel variants, four of them classified as pathogenic. In one family, we identified two mutations in exon 102, segregating in cis, suggesting an additive effect of two mutations in the same allele. This work highlights the importance of using Next-Generation Sequencing technology for the molecular diagnosis of genetic diseases when a very large gene is involved, associated to a broad distribution of the mutations along it. These data also influence the prevention through adequate genetic counseling for the families and cautions against malignant hyperthermia susceptibility.
- Published
- 2020
43. Central Core Disease: Facial Weakness Differentiating Biallelic from Monoallelic Forms
- Author
-
Ana Cotta, Lucas Santos Souza, Elmano Carvalho, Leticia Nogueira Feitosa, Antonio Cunha, Monica Machado Navarro, Jaquelin Valicek, Miriam Melo Menezes, Simone Vilela Nunes Neves, Rafael Xavier-Neto, Antonio Pedro Vargas, Reinaldo Issao Takata, Julia Filardi Paim, and Mariz Vainzof
- Subjects
Neuroblastoma ,Genetics ,High-Throughput Nucleotide Sequencing ,Humans ,Ryanodine Receptor Calcium Release Channel ,Myopathy, Central Core ,central core disease ,RYR1 ,clinical heterogeneity ,electromyography ,Genetics (clinical) ,Pedigree ,Retrospective Studies - Abstract
Central Core Disease (CCD) is a genetic neuromuscular disorder characterized by the presence of cores in muscle biopsy. The inheritance has been described as predominantly autosomal dominant (AD), and the disease may present as severe neonatal or mild adult forms. Here we report clinical and molecular data on a large cohort of Brazilian CCD patients, including a retrospective clinical analysis and molecular screening for RYR1 variants using Next-Generation Sequencing (NGS). We analyzed 27 patients from 19 unrelated families: four families (11 patients) with autosomal dominant inheritance (AD), two families (3 patients) with autosomal recessive (AR), and 13 sporadic cases. Biallelic RYR1 variants were found in six families (two AR and four sporadic cases) of the 14 molecularly analyzed families (~43%), suggesting a higher frequency of AR inheritance than expected. None of these cases presented a severe phenotype. Facial weakness was more common in biallelic than in monoallelic patients (p = 0.0043) and might be a marker for AR forms. NGS is highly effective for the identification of RYR1 variants in CCD patients, allowing the discovery of a higher proportion of AR cases with biallelic mutations. These data have important implications for the genetic counseling of the families.
- Published
- 2022
44. Ryanodine receptor 1-related disorders: an historical perspective and proposal for a unified nomenclature
- Author
-
James J. Dowling, Robert T. Dirksen, Jessica W Witherspoon, Joshua J. Todd, Katherine G. Meilleur, Tokunbor A. Lawal, Susan L. Hamilton, and Carsten G. Bönnemann
- Subjects
0301 basic medicine ,myalgia ,Pathology ,medicine.medical_specialty ,History ,Neuromuscular disease ,lcsh:Diseases of the musculoskeletal system ,Myopathy ,Skeletal muscle ,Review ,03 medical and health sciences ,0302 clinical medicine ,Ion channel defects ,Terminology as Topic ,medicine ,Animals ,Humans ,Orthopedics and Sports Medicine ,Molecular Biology ,RYR1 ,Ryanodine receptor ,business.industry ,Malignant hyperthermia ,Ryanodine Receptor Calcium Release Channel ,Periodic paralysis ,Neuromuscular Diseases ,Cell Biology ,medicine.disease ,musculoskeletal system ,Phenotype ,030104 developmental biology ,Clinical neurology ,medicine.symptom ,lcsh:RC925-935 ,business ,tissues ,030217 neurology & neurosurgery ,Central core disease - Abstract
TheRYR1gene, which encodes the sarcoplasmic reticulum calcium release channel or type 1 ryanodine receptor (RyR1) of skeletal muscle, was sequenced in 1988 andRYR1variations that impair calcium homeostasis and increase susceptibility to malignant hyperthermia were first identified in 1991. Since then,RYR1-related myopathies (RYR1-RM) have been described as rare, histopathologically and clinically heterogeneous, and slowly progressive neuromuscular disorders.RYR1variants can lead to dysfunctional RyR1-mediated calcium release, malignant hyperthermia susceptibility, elevated oxidative stress, deleterious post-translational modifications, and decreased RyR1 expression.RYR1-RM-affected individuals can present with delayed motor milestones, contractures, scoliosis, ophthalmoplegia, and respiratory insufficiency.Historically,RYR1-RM-affected individuals were diagnosed based on morphologic features observed in muscle biopsies including central cores, cores and rods, central nuclei, fiber type disproportion, and multi-minicores. However, these histopathologic features are not always specific toRYR1-RM and often change over time. As additional phenotypes were associated withRYR1variations (including King-Denborough syndrome, exercise-induced rhabdomyolysis, lethal multiple pterygium syndrome, adult-onset distal myopathy, atypical periodic paralysis with or without myalgia, mild calf-predominant myopathy, and dusty core disease) the overlap among diagnostic categories is ever increasing. With the continuing emergence of new clinical subtypes along theRYR1disease spectrum and reports of adult-onset phenotypes, nuanced nomenclatures have been reported (RYR1- [related, related congenital, congenital] myopathies). In this narrative review, we provide historical highlights ofRYR1research, accounts of the main diagnostic disease subtypes and proposeRYR1-related disorders (RYR1-RD) as a unified nomenclature to describe this complex and evolving disease spectrum.
- Published
- 2020
- Full Text
- View/download PDF
45. New Compound Heterozygous Splice Site Mutations of the Skeletal Muscle Ryanodine Receptor (RYR1) Gene Manifest Fetal Akinesia: A Linkage with Congenital Myopathies
- Author
-
Ioannis Tsakiridis, Themistoklis Dagklis, Tryfon Tsagas, Anastasios Sartsidis, Makarios Eleftheriades, Milica Filipovic Stankovic, Georgios Kalogeros, Ioannis Papoulidis, Ioannis Tziotis, Vladimir Arsenijevic, Nebojsa Zecevic, Emmanouil Manolakos, and Georgios Theocharis
- Subjects
RYR1 ,0303 health sciences ,medicine.medical_specialty ,Fetus ,Splice site mutation ,030305 genetics & heredity ,Malignant hyperthermia ,Biology ,musculoskeletal system ,medicine.disease ,Compound heterozygosity ,03 medical and health sciences ,Endocrinology ,Internal medicine ,Genetics ,medicine ,splice ,Genetics (clinical) ,Central core disease ,Minicore myopathy ,030304 developmental biology - Abstract
Mutations in the skeletal muscle ryanodine receptor (RYR1) gene have been linked to malignant hyperthermia susceptibility, central core disease, and minicore myopathy with external ophthalmoplegia. RYR1 is an intracellular calcium release channel and plays a crucial role in the sarcoplasmic reticulum and transverse tubule connection. Here, we report 2 fetuses from the same parents with compound heterozygous mutations in the RYR1 gene (c.10347+1G>A and c.10456-2Α>G) who presented with fetal akinesia and polyhydramnios at 27 and 19 weeks of gestation with intrauterine growth restriction in the third pregnancy. The prospective parents of the fetuses were heterozygous carriers for c.10456-2Α>G (mother) and c.10347+1G>A (father). Both mutations affect splice sites resulting in dysfunctional protein forms probably missing crucial domains of the C-terminus. Our findings reveal a new RYR1 splice site mutation (c.10456-2Α>G) that may be associated with the clinical features of myopathies, expanding the RYR1 spectrum related to these pathologies.
- Published
- 2020
- Full Text
- View/download PDF
46. Significance of Asymptomatic Hyper Creatine-Kinase Emia
- Author
-
Fulvio A. Scorza, Carla A. Scorza, and Josef Finsterer
- Subjects
0301 basic medicine ,Dysferlinopathy ,Pediatrics ,medicine.medical_specialty ,Mitochondrial Diseases ,030105 genetics & heredity ,Asymptomatic ,03 medical and health sciences ,0302 clinical medicine ,Muscular Diseases ,medicine ,Humans ,Mass Screening ,Family history ,Myopathy ,Creatine Kinase ,Mass screening ,Muscle biopsy ,medicine.diagnostic_test ,business.industry ,Malignant hyperthermia ,General Medicine ,medicine.disease ,Neurology ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Central core disease - Abstract
Objectives Whether asymptomatic hyper-CKemia (AHCE) should prompt a thorough work-up for muscle disease or not is controversially discussed. This review aims at summarizing and discussing recent findings concerning the cause, frequency, evolution, and work-up of conditions manifesting as AHCE and normal or abnormal electromyography (EMG) respectively muscle biopsy. Methods Systematic PubMed search. Results There are numerous primary (hereditary) and acquired myopathies that manifest with permanent, recurrent, or temporary AHCE with/without myopathic EMG or muscle biopsy. AHCE particularly occurs at onset of these conditions, which include dystrophinopathies, myotilinopathies, calpainopathy, caveolinopathy, dysferlinopathy, central core disease, multicore disease, desminopathy, MD1, MD2, hypoPP, malignant hyperthermia susceptibility, Pompe disease, McArdle disease, myoadenylate deaminase-deficiency, CPT2-deficiency, mitochondrial disorders, or myopathy with tubular aggregates. Most likely, other primary myopathies manifest with AHCE as well, without having been reported. Patients with AHCE should be taken seriously and repeated CK determination must be conducted. If hyper-CKemia is persisting or recurrent, these patients should undergo an EMG and eventually muscle biopsy. If noninformative, genetic work-up by a panel or whole exome sequencing should be initiated, irrespective of the family history. Patients with AHCE should avoid excessive exercise, require sufficient hydration, require counseling with regard to the risk of malignant hyperthermia, and should inform anesthesiologists and surgeons about their condition before elective surgery. Conclusions Recurrent AHCE should be taken seriously and managed with conventional work-up. If noninformative, genetic work-up should follow irrespective of the family history.
- Published
- 2019
- Full Text
- View/download PDF
47. A recurrent RYR1 mutation associated with early-onset hypotonia and benign disease course
- Author
-
Julie Perrier-Boeswillwald, Johann Böhm, Eric Bieth, Marie-Christine Minot-Myhie, Marie-Christine Nougues, Annabelle Chaussenot, Helen Mecili, François-Jérôme Authier, Maud Michaud, Sandra Mercier, Norma B. Romero, Claude Cances, Julien Fauré, Mégane Pizzimenti, Nicolas Dondaine, Valérie Biancalana, Sabrina Sacconi, Antoinette Gelot Bernabe, John Rendu, Alison Bouzenard, Armelle Magot, Bertrand Isidor, Yann Péréon, Mélanie Fradin, Emmanuelle Uro-Coste, Jocelyn Laporte, Pascale Marcorelles, Gilles Bretaudeau, Laurent Pasquier, Ana Ferreiro, Bruno Eymard, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), CHU Strasbourg, Centre Hospitalier Universitaire [Grenoble] (CHU), [GIN] Grenoble Institut des Neurosciences (GIN), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes (UGA), Hôpital l'Archet, Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Service de Génétique [Purpan], CHU Toulouse [Toulouse], Hôpital Sud [CHU Rennes], CHU Pontchaillou [Rennes], Centre de référence Maladies Rares CLAD-Ouest [Rennes], Centre hospitalier universitaire de Nantes (CHU Nantes), Reference Centre for Neuromuscular Disorders ( FILNEMUS), CHU Trousseau [APHP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Centre Hospitalier Universitaire de Nice (CHU Nice), Université Côte d'Azur (UCA), Institut de Myologie, Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Association française contre les myopathies (AFM-Téléthon)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), CHU Pitié-Salpêtrière [AP-HP], Centre Hospitalier Régional Universitaire de Brest (CHRU Brest), Université de Bretagne Occidentale - UFR Médecine et Sciences de la Santé (UBO UFR MSS), Université de Brest (UBO), Hôpital Henri Mondor, Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Institut de Neurobiologie de la Méditerranée [Aix-Marseille Université] (INMED - INSERM U1249), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Unité de Biologie Fonctionnelle et Adaptative (BFA (UMR_8251 / U1133)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Dynamique et Structure du Cytosquelette Neuronal, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes (UGA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Grenoble Alpes (UGA), Service Génétique Médicale [CHU Toulouse], Institut Fédératif de Biologie (IFB), Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Pôle Biologie [CHU Toulouse], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), and Laporte, Jocelyn
- Subjects
Male ,Pathology ,Triad ,Neuromuscular disorder ,Case Report ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,[SDV.BBM.BM] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,0302 clinical medicine ,Age of Onset ,0303 health sciences ,Congenital myopathy ,medicine.diagnostic_test ,Muscle weakness ,Middle Aged ,musculoskeletal system ,Hypotonia ,3. Good health ,Pedigree ,[SDV.SP.PHARMA] Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Child, Preschool ,Disease Progression ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Muscle Hypotonia ,Female ,medicine.symptom ,Adult ,medicine.medical_specialty ,Adolescent ,[SDV.GEN.GA] Life Sciences [q-bio]/Genetics/Animal genetics ,Muscle disorder ,[SDV.GEN.GH] Life Sciences [q-bio]/Genetics/Human genetics ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,Excitation–contraction coupling ,[SDV.MHEP.PED] Life Sciences [q-bio]/Human health and pathology/Pediatrics ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,medicine ,[SDV.BC.BC] Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Humans ,Centronuclear myopathy ,RC346-429 ,030304 developmental biology ,Aged ,RYR1 ,[SDV.MHEP.PED]Life Sciences [q-bio]/Human health and pathology/Pediatrics ,Muscle biopsy ,business.industry ,Ryanodine Receptor Calcium Release Channel ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,medicine.disease ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Calcium ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,business ,030217 neurology & neurosurgery ,Central core disease - Abstract
The ryanodine receptor RyR1 is the main sarcoplasmic reticulum Ca2+ channel in skeletal muscle and acts as a connecting link between electrical stimulation and Ca2+-dependent muscle contraction. Abnormal RyR1 activity compromises normal muscle function and results in various human disorders including malignant hyperthermia, central core disease, and centronuclear myopathy. However, RYR1 is one of the largest genes of the human genome and accumulates numerous missense variants of uncertain significance (VUS), precluding an efficient molecular diagnosis for many patients and families. Here we describe a recurrent RYR1 mutation previously classified as VUS, and we provide clinical, histological, and genetic data supporting its pathogenicity. The heterozygous c.12083C>T (p.Ser4028Leu) mutation was found in thirteen patients from nine unrelated congenital myopathy families with consistent clinical presentation, and either segregated with the disease in the dominant families or occurred de novo. The affected individuals essentially manifested neonatal or infancy-onset hypotonia, delayed motor milestones, and a benign disease course differing from classical RYR1-related muscle disorders. Muscle biopsies showed unspecific histological and ultrastructural findings, while RYR1-typical cores and internal nuclei were seen only in single patients. In conclusion, our data evidence the causality of the RYR1 c.12083C>T (p.Ser4028Leu) mutation in the development of an atypical congenital myopathy with gradually improving motor function over the first decades of life, and may direct molecular diagnosis for patients with comparable clinical presentation and unspecific histopathological features on the muscle biopsy.
- Published
- 2021
- Full Text
- View/download PDF
48. Chromosome 17 Candidate Gene Analysis in a Population Referred Because of Suspected Susceptibility to Malignant Hyperthermia
- Author
-
Vita, Gary M., Olckers, Antonel, Jedlicka, Anne E., Heiman-Patterson, Terry, George, Alfred L., Fletcher, Jeffery E., Rosenberg, Henry, Levitt, Roy C., Morio, Michio, editor, Kikuchi, Hirosato, editor, and Yuge, Osafumi, editor
- Published
- 1996
- Full Text
- View/download PDF
49. RYR1-related myopathies: Expanding the spectrum of morphological presentation
- Author
-
Vincenzo Sorrentino
- Subjects
Genetics ,RYR1 ,Physiology ,Malignant hyperthermia ,Biology ,Muscle disorder ,musculoskeletal system ,medicine.disease ,Congenital myopathy ,Phenotype ,medicine ,tissues ,Gene ,Central core disease ,Pharmacogenetics - Abstract
Mutations in the RYR1 gene are the most common cause of nondystrophic congenital myopathies. Mutations in RYR1 were initially identified in individuals susceptible to malignant hyperthermia, a pharmacogenetic disorder triggered by volatile anesthetics and succinylcholine. Shortly after, mutations in RYR1 were identified in patients with central core disease, which is the most frequent congenital myopathy, and in other muscle disorders, collectively referred to as RYR1-related myopathies. RYR1 mutations are also responsible of some acute pathological conditions triggered by heat- and exercise-induced stress, named exertional heat stroke and exertional-induced rhabdomyolysis, which, similarly to malignant hyperthermia, occur in otherwise healthy individuals with normal skeletal muscle functions. Hundreds of causative mutations linked to RYR1-related diseases have been identified. These mutations are clustered in three regions that are referred to as the N-terminal, central, and C-terminal hot spots. Recent developments in cryo-EM techniques have provided high-resolution reconstructions of the channel, allowing a much better definition of the structural domains within the large N-terminal cytoplasmic region and in the C-terminal domain containing six transmembrane helices and the pore region of the channel. RYR1 mutations may either activate or inhibit channel function or, in some cases, can reduce the expression levels of RYR1 protein. However, similar clinical phenotypes can result from mutations with opposing effects on RYR1 function, or little or no correlation can be found between the observed clinical phenotype and localization of mutations in the structural domains of the RYR1 channel, even though recent studies indicate that clinically severe cases are mostly recessive or due to mutations located in the bridging solenoid. Recent results on the identification of RYR1 mutations in patients with myopathies will be presented.
- Published
- 2021
- Full Text
- View/download PDF
50. Molecular basis of impaired muscle function in a mouse model of congenital myopathy due to compound heterozygous RYR1 mutations
- Author
-
Alexis Ruizl, F Zorzato, Jan Eckhardt, and Susan Treves
- Subjects
RYR1 ,Pathology ,medicine.medical_specialty ,Muscle biopsy ,medicine.diagnostic_test ,Physiology ,Muscle weakness ,Muscle disorder ,Biology ,musculoskeletal system ,medicine.disease ,Compound heterozygosity ,Congenital myopathy ,medicine ,Centronuclear myopathy ,medicine.symptom ,Central core disease - Abstract
Congenital myopathies (CM) are a group of early-onset, genetically diverse muscle disorders of variable severity with characteristic muscle biopsy findings. Mutations in RYR1, the gene encoding the RYR1, are the most common genetic cause, responsible for ∼30% of all human CM. They are linked to the pharmacogenetic disorder malignant hyperthermia susceptibility and to various disease phenotypes, including central core disease (which is primarily dominantly inherited), multiminicore disease (which is predominantly recessively inherited), some forms of centronuclear myopathy and congenital fiber-type disproportion (which can be either dominantly or recessively inherited), and King–Denborough syndrome (a CM characterized by skeletal abnormalities, dysmorphic features, and malignant hyperthermia susceptibility). The recessive forms of RYR1-linked CM are more severe, affecting children at birth and, in addition to profound muscle weakness, may also affect facial and extraocular muscles and cause skeletal deformities and feeding difficulties. To study the mechanism leading to the profound muscle weakness characterized by recessive RYR1-CM, we created transgenic mice knocked in for the compound heterozygous RYR1 p.Q1970fsX16+p.A4329D mutations (double knock-in mouse, or DKI) identified in a severely affected child. The in vivo and ex vivo physiological functions of fast twitch, slow twitch, and extraocular muscles were severely impaired in DKI mice; in addition, the mutations were accompanied by a >50% decrease in RYR1 protein in all muscles examined, as well as changes in the expression of many proteins important for muscle function and chromatin structure. Muscle ultrastructure was disorganized, with fewer CRU and mitochondria and presence of cores. MyHC-EO, the superfast and ocular-muscle−specific myosin heavy isoform, was almost undetectable in EOMs from DKI mutant mice. Thus, the DKI mouse model faithfully recapitulates the human disease and could be exploited for preclinical studies aimed at developing therapeutic strategies to treat neuromuscular disorders linked to recessive RYR1 mutations.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.