1
|
Bang I, Hattori T, Leloup N, Corrado A, Nyamaa A, Koide A, Geles K, Buck E, Koide S. Selective targeting of oncogenic hotspot mutations of the HER2 extracellular domain. Nat Chem Biol 2024:10.1038/s41589-024-01751-w. [PMID: 39438724 DOI: 10.1038/s41589-024-01751-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
Oncogenic mutations in the extracellular domain (ECD) of cell-surface receptors could serve as tumor-specific antigens that are accessible to antibody therapeutics. Such mutations have been identified in receptor tyrosine kinases including HER2. However, it is challenging to selectively target a point mutant, while sparing the wild-type protein. Here we developed antibodies selective to HER2 S310F and S310Y, the two most common oncogenic mutations in the HER2 ECD, via combinatorial library screening and structure-guided design. Cryogenic-electron microscopy structures of the HER2 S310F homodimer and an antibody bound to HER2 S310F revealed that these antibodies recognize the mutations in a manner that mimics the dimerization arm of HER2 and thus inhibit HER2 dimerization. These antibodies as T cell engagers selectively killed a HER2 S310F-driven cancer cell line in vitro, and in vivo as a xenograft. These results validate HER2 ECD mutations as actionable therapeutic targets and offer promising candidates toward clinical development.
Collapse
Affiliation(s)
- Injin Bang
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Takamitsu Hattori
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nadia Leloup
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Alexis Corrado
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Atekana Nyamaa
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Akiko Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Division of Hematology Oncology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Ken Geles
- Black Diamond Therapeutics, New York, NY, USA
| | | | - Shohei Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Ramdas S, Beeson D, Dong YY. Congenital myasthenic syndromes: increasingly complex. Curr Opin Neurol 2024; 37:493-501. [PMID: 39051439 PMCID: PMC11377046 DOI: 10.1097/wco.0000000000001300] [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: 07/27/2024]
Abstract
PURPOSE OF REVIEW Congenital myasthenia syndromes (CMS) are treatable, inherited disorders affecting neuromuscular transmission. We highlight that the involvement of an increasing number of proteins is making the understanding of the disease mechanisms and potential treatments progressively more complex. RECENT FINDINGS Although early studies identified mutations of proteins directly involved in synaptic transmission at the neuromuscular junction, recently, next-generation sequencing has facilitated the identification of many novel mutations in genes that encode proteins that have a far wider expression profile, some even ubiquitously expressed, but whose defective function leads to impaired neuromuscular transmission. Unsurprisingly, mutations in these genes often causes a wider phenotypic disease spectrum where defective neuromuscular transmission forms only one component. This has implications for the management of CMS patients. SUMMARY Given the widening nonneuromuscular junction phenotypes in the newly identified forms of CMS, new therapies need to include disease-modifying approaches that address not only neuromuscular weakness but also the multisystem involvement. Whilst the current treatments for CMS are highly effective for many subtypes there remains, in a proportion of CMS patients, an unmet need for more efficacious therapies.
Collapse
Affiliation(s)
- Sithara Ramdas
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford
- Department of Paediatric Neurology, John Radcliffe Hospital
| | - David Beeson
- Neurosciences Group, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford OX3 9DS
| | - Yin Yao Dong
- Neurosciences Group, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford OX3 9DS
| |
Collapse
|
3
|
Oury J, Gamallo-Lana B, Santana L, Steyaert C, Vergoossen DLE, Mar AC, Vankerckhoven B, Silence K, Vanhauwaert R, Huijbers MG, Burden SJ. Agonist antibody to MuSK protects mice from MuSK myasthenia gravis. Proc Natl Acad Sci U S A 2024; 121:e2408324121. [PMID: 39288173 PMCID: PMC11441477 DOI: 10.1073/pnas.2408324121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Myasthenia gravis (MG) is a chronic and severe disease of the skeletal neuromuscular junction (NMJ) in which the effects of neurotransmitters are attenuated, leading to muscle weakness. In the most common forms of autoimmune MG, antibodies attack components of the postsynaptic membrane, including the acetylcholine receptor (AChR) or muscle-specific kinase (MuSK). MuSK, a master regulator of NMJ development, associates with the low-density lipoprotein-related receptor 4 (Lrp4) to form the signaling receptor for neuronal Agrin, a nerve-derived synaptic organizer. Pathogenic antibodies to MuSK interfere with binding between MuSK and Lrp4, inhibiting the differentiation and maintenance of the NMJ. MuSK MG can be debilitating and refractory to treatments that are effective for AChR MG. We show here that recombinant antibodies, derived from MuSK MG patients, cause severe neuromuscular disease in mice. The disease can be prevented by a MuSK agonist antibody, presented either prophylactically or after disease onset. These findings suggest a therapeutic alternative to generalized immunosuppression for treating MuSK MG by selectively and directly targeting the disease mechanism.
Collapse
Affiliation(s)
- Julien Oury
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Medical School, New York, NY10016
| | - Begona Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University School of Medicine, New York, NY10016
| | - Leah Santana
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Medical School, New York, NY10016
| | | | - Dana L. E. Vergoossen
- Department of Human Genetics, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
- Department of Neurology, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
| | - Adam C. Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University School of Medicine, New York, NY10016
| | | | | | | | - Maartje G. Huijbers
- Department of Human Genetics, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
- Department of Neurology, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
| | - Steven J. Burden
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Medical School, New York, NY10016
| |
Collapse
|
4
|
Vanhauwaert R, Oury J, Vankerckhoven B, Steyaert C, Jensen SM, Vergoossen DLE, Kneip C, Santana L, Lim JL, Plomp JJ, Augustinus R, Koide S, Blanchetot C, Ulrichts P, Huijbers MG, Silence K, Burden SJ. ARGX-119 is an agonist antibody for human MuSK that reverses disease relapse in a mouse model of congenital myasthenic syndrome. Sci Transl Med 2024; 16:eado7189. [PMID: 39292800 DOI: 10.1126/scitranslmed.ado7189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024]
Abstract
Muscle-specific kinase (MuSK) is essential for the formation, function, and preservation of neuromuscular synapses. Activation of MuSK by a MuSK agonist antibody may stabilize or improve the function of the neuromuscular junction (NMJ) in patients with disorders of the NMJ, such as congenital myasthenia (CM). Here, we generated and characterized ARGX-119, a first-in-class humanized agonist monoclonal antibody specific for MuSK, that is being developed for treatment of patients with neuromuscular diseases. We performed in vitro ligand-binding assays to show that ARGX-119 binds with high affinity to the Frizzled-like domain of human, nonhuman primate, rat, and mouse MuSK, without off-target binding, making it suitable for clinical development. Within the Fc region, ARGX-119 harbors L234A and L235A mutations to diminish potential immune-activating effector functions. Its mode of action is to activate MuSK, without interfering with its natural ligand neural Agrin, and cluster acetylcholine receptors in a dose-dependent manner, thereby stabilizing neuromuscular function. In a mouse model of DOK7 CM, ARGX-119 prevented early postnatal lethality and reversed disease relapse in adult Dok7 CM mice by restoring neuromuscular function and reducing muscle weakness and fatigability in a dose-dependent manner. Pharmacokinetic studies in nonhuman primates, rats, and mice revealed a nonlinear PK behavior of ARGX-119, indicative of target-mediated drug disposition and in vivo target engagement. On the basis of this proof-of-concept study, ARGX-119 has the potential to alleviate neuromuscular diseases hallmarked by impaired neuromuscular synaptic function, warranting further clinical development.
Collapse
Affiliation(s)
| | - Julien Oury
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | | | | | - Stine Marie Jensen
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Dana L E Vergoossen
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | | | - Leah Santana
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Jamie L Lim
- argenx, 9052 Zwijnaarde, Belgium
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Jaap J Plomp
- Department of Neurology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Roy Augustinus
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Shohei Koide
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | | | | | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, Netherlands
- Department of Neurology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | | | - Steven J Burden
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
5
|
Jin M, Xie M, Dong L, Xue F, Li W, Jiang L, Li J, Zhang M, Song H, Lu Q, Yu Q. Exploration of Positive and Negative Schizophrenia Symptom Heterogeneity and Establishment of Symptom-Related miRNA-mRNA Regulatory Network: Based on Transcriptome Sequencing Data. Mol Neurobiol 2024; 61:5992-6012. [PMID: 38267752 DOI: 10.1007/s12035-024-03942-x] [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: 04/27/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024]
Abstract
Schizophrenia (SCZ) symptoms can be classified as positive and negative ones, each of which has distinct traits and possibly differences in gene expression and regulation. The co-expression networks linked to PANSS (Positive and Negative Syndrome Scale) scores were identified by weighted gene co-expression network analysis (WGCNA) using the expression profiles of miRNA and mRNA in the peripheral blood of first-episode SCZ patients. The heterogeneity between positive and negative symptoms was demonstrated using gene functional enrichment, gene-medication interaction, and immune cell composition analysis. Then, target gene prediction and correlation analysis of miRNA and mRNA constructed a symptom-related miRNA-mRNA regulatory network, screened regulatory pairs, and predicted binding sites. A total of six mRNA co-expression modules, two miRNA co-expression modules, and ten hub genes were screened to be significantly associated with positive symptoms; five mRNA co-expression modules and eight hub genes were correlated with negative symptoms. Positive symptom-related modules were significantly enriched in axon guidance, actin skeleton regulation, and sphingolipid signaling pathway, while negative symptom-related modules were significantly enriched in adaptive immune response, leukocyte migration, dopaminergic synapses, etc. The development of positive symptoms may have been influenced by potential regulatory pairings such as miR-98-5p-EIF3J, miR-98-5p-SOCS4, let-7b-5p-CLUH, miR-454-3p-GTF2H1, and let-7b-5p-SNX17. Additionally, immune cells were substantially connected with several hub genes for symptoms. Positive and negative symptoms in SCZ individuals were heterogeneous to some extent. miRNAs such as let-7b-5p and miR-98-5p might contribute to the incidence of positive symptoms by targeting mRNAs, while the immune system's role in developing negative symptoms may be more nuanced.
Collapse
Affiliation(s)
- Mengdi Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Mengtong Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Lin Dong
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Fengyu Xue
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Weizhen Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Lintong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Junnan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Min Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Haideng Song
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Qingxing Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, China.
| |
Collapse
|
6
|
Cocanougher BT, Liu SW, Francescatto L, Behura A, Anneling M, Jackson DG, Deak KL, Hornik CD, ElMallah MK, Pizoli CE, Smith EC, Tan KGQ, McDonald MT. The severity of MUSK pathogenic variants is predicted by the protein domain they disrupt. HGG ADVANCES 2024; 5:100288. [PMID: 38566418 PMCID: PMC11070630 DOI: 10.1016/j.xhgg.2024.100288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Biallelic loss-of-function variants in the MUSK gene result in two allelic disorders: (1) congenital myasthenic syndrome (CMS; OMIM: 616325), a neuromuscular disorder that has a range of severity from severe neonatal-onset weakness to mild adult-onset weakness, and (2) fetal akinesia deformation sequence (OMIM: 208150), a form of pregnancy loss characterized by severe muscle weakness in the fetus. The MUSK gene codes for muscle-specific kinase (MuSK), a receptor tyrosine kinase involved in the development of the neuromuscular junction. Here, we report a case of neonatal-onset MUSK-related CMS in a patient harboring compound heterozygous deletions in the MUSK gene, including (1) a deletion of exons 2-3 leading to an in-frame MuSK protein lacking the immunoglobulin 1 (Ig1) domain and (2) a deletion of exons 7-11 leading to an out-of-frame, truncated MuSK protein. Individual domains of the MuSK protein have been elucidated structurally; however, a complete MuSK structure generated by machine learning algorithms has clear inaccuracies. We modify a predicted AlphaFold structure and integrate previously reported domain-specific structural data to suggest a MuSK protein that dimerizes in two locations (Ig1 and the transmembrane domain). We analyze known pathogenic variants in MUSK to discover domain-specific genotype-phenotype correlations; variants that lead to a loss of protein expression, disruption of the Ig1 domain, or Dok-7 binding are associated with the most severe phenotypes. A conceptual model is provided to explain the severe phenotypes seen in Ig1 variants and the poor response of our patient to pyridostigmine.
Collapse
Affiliation(s)
- Benjamin T Cocanougher
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Medical Genetics, Duke University, Durham, NC, USA.
| | - Samuel W Liu
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Medical Genetics, Duke University, Durham, NC, USA
| | | | - Alexander Behura
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Mariele Anneling
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Medical Genetics, Duke University, Durham, NC, USA
| | - David G Jackson
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Kristen L Deak
- Department of Pathology, Duke University, Durham, NC, USA
| | - Chi D Hornik
- Department of Pediatrics, Duke University, Durham, NC, USA
| | - Mai K ElMallah
- Department of Pediatrics, Duke University, Durham, NC, USA
| | - Carolyn E Pizoli
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Pediatric Neurology, Duke University, Durham, NC, USA
| | - Edward C Smith
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Pediatric Neurology, Duke University, Durham, NC, USA
| | - Khoon Ghee Queenie Tan
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Marie T McDonald
- Department of Pediatrics, Duke University, Durham, NC, USA; Division of Medical Genetics, Duke University, Durham, NC, USA.
| |
Collapse
|
7
|
Sun S, Shen Y, Zhang X, Ding N, Xu Z, Zhang Q, Li L. The MuSK agonist antibody protects the neuromuscular junction and extends the lifespan in C9orf72-ALS mice. Mol Ther 2024; 32:2176-2189. [PMID: 38734896 PMCID: PMC11286808 DOI: 10.1016/j.ymthe.2024.05.016] [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: 11/08/2023] [Revised: 04/06/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
The disassembly of the neuromuscular junction (NMJ) is an early event in amyotrophic lateral sclerosis (ALS), ultimately leading to motor dysfunction and lethal respiratory paralysis. The hexanucleotide GGGGCC repeat expansion in the C9orf72 gene is the most common genetic mutation, and the dipeptide repeat (DPR) proteins have been shown to cause neurodegeneration. While no drugs can treat ALS patients efficiently, new treatment strategies are urgently needed. Here, we report that a MuSK agonist antibody alleviates poly-PR-induced NMJ deficits in C9orf72-ALS mice. The HB9-PRF/F mice, which express poly-PR proteins in motor neurons, exhibited impaired motor behavior and NMJ deficits. Mechanistically, poly-PR proteins interacted with Agrin to disrupt the interaction between Agrin and Lrp4, leading to attenuated activation of MuSK. Treatment with a MuSK agonist antibody rescued NMJ deficits, and extended the lifespan of C9orf72-ALS mice. Moreover, impaired NMJ transmission was observed in C9orf72-ALS patients. These findings identify the mechanism by which poly-PR proteins attenuate MuSK activation and NMJ transmission, highlighting the potential of promoting MuSK activation with an agonist antibody as a therapeutic strategy to protect NMJ function and prolong the lifespan of ALS patients.
Collapse
Affiliation(s)
- Shuangshuang Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yihui Shen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xu Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ning Ding
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhe Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qijie Zhang
- Department of Neurology, Fujian Institute of Neurology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.
| | - Lei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
8
|
Ziaadini B, Ghaderi Yazdi B, Dirandeh E, Boostani R, Karimi N, Panahi A, Kariminejad A, Fadaee M, Ahangari F, Nafissi S. DOK7 congenital myasthenic syndrome: case series and review of literature. BMC Neurol 2024; 24:211. [PMID: 38907197 PMCID: PMC11191154 DOI: 10.1186/s12883-024-03713-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/05/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Congenital myasthenic syndromes (CMS) are among the most challenging differential diagnoses in the neuromuscular domain, consisting of diverse genotypes and phenotypes. A mutation in the Docking Protein 7 (Dok-7) is a common cause of CMS. DOK7 CMS requires different treatment than other CMS types. Regarding DOK7's special considerations and challenges ahead of neurologists, we describe seven DOK7 patients and evaluate their response to treatment. METHODS The authors visited these patients in the neuromuscular clinics of Tehran and Kerman Universities of Medical Sciences Hospitals. They diagnosed these patients based on clinical findings and neurophysiological studies, which Whole Exome Sequencing confirmed. For each patient, we tried unique medications and recorded the clinical response. RESULTS The symptoms started from birth to as late as the age of 33, with the mean age of onset being 12.5. Common symptoms were: Limb-girdle weakness in 6, fluctuating symptoms in 5, ptosis in 4, bifacial weakness in 3, reduced extraocular movement in 3, bulbar symptoms in 2 and dyspnea in 2 3-Hz RNS was decremental in 5 out of 6 patients. Salbutamol was the most effective. c.1124_1127dupTGCC is the most common variant; three patients had this variant. CONCLUSION We strongly recommend that neurologists consider CMS in patients with these symptoms and a similar familial history. We recommend prescribing salbutamol as the first-choice treatment option for DOK7 patients.
Collapse
Affiliation(s)
- Bentolhoda Ziaadini
- Neurology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Bardyia Ghaderi Yazdi
- Neuromuscular Research Center, Shariati Hospital, Tehran University of Medical Sciences, Shariati Hospital, North Karegar St, Tehran, 14117-13135, Iran
- Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Dirandeh
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan, Iran
| | - Reza Boostani
- Department of Neurology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Karimi
- Department of Neurology, School of Medicine, Immunogenetics Research Center, Toxoplasmosis Research Center, Clinical Research Development Unit of Bou Ali Sina Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Akram Panahi
- Neuromuscular Research Center, Shariati Hospital, Tehran University of Medical Sciences, Shariati Hospital, North Karegar St, Tehran, 14117-13135, Iran
- Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mahsa Fadaee
- Kariminejad-Najmabadi Pathology & Genetics Center, Tehran, Iran
| | | | - Shahriar Nafissi
- Neuromuscular Research Center, Shariati Hospital, Tehran University of Medical Sciences, Shariati Hospital, North Karegar St, Tehran, 14117-13135, Iran.
- Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Herbst R, Huijbers MG, Oury J, Burden SJ. Building, Breaking, and Repairing Neuromuscular Synapses. Cold Spring Harb Perspect Biol 2024; 16:a041490. [PMID: 38697654 PMCID: PMC11065174 DOI: 10.1101/cshperspect.a041490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
A coordinated and complex interplay of signals between motor neurons, skeletal muscle cells, and Schwann cells controls the formation and maintenance of neuromuscular synapses. Deficits in the signaling pathway for building synapses, caused by mutations in critical genes or autoantibodies against key proteins, are responsible for several neuromuscular diseases, which cause muscle weakness and fatigue. Here, we describe the role that four key genes, Agrin, Lrp4, MuSK, and Dok7, play in this signaling pathway, how an understanding of their mechanisms of action has led to an understanding of several neuromuscular diseases, and how this knowledge has contributed to emerging therapies for treating neuromuscular diseases.
Collapse
Affiliation(s)
- Ruth Herbst
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Centre LUMC, 2300 RC Leiden, the Netherlands
- Department of Neurology, Leiden University Medical Centre LUMC, 2333 ZA Leiden, the Netherlands
| | - Julien Oury
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, New York 10016, USA
| | - Steven J Burden
- Neurology Department, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
10
|
Keane SP, Chadman KK, Gomez AR, Hu W. Pros and cons of narrow- versus wide-compartment rotarod apparatus: An experimental study in mice. Behav Brain Res 2024; 463:114901. [PMID: 38341101 DOI: 10.1016/j.bbr.2024.114901] [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: 11/02/2023] [Revised: 01/23/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
The rotarod test, a sensorimotor assessment that allows for quantitative evaluation of motor coordination in rodents, has extensive application in many research fields. The test results exhibit extreme between-study variability, sometimes making it challenging to conclude the validity of certain disease models and related therapeutic effects. Although the variation in test paradigms may account for this disparity, some features of rotarod apparatus including rod diameter make differences. However, it is unknown whether the width of animal compartment has a role in rotarod performance. Here we comprehensively evaluated the active rotarod performance and adverse incidents in multiple strains of mice on an 11-cm- or a 5-cm-wide compartment apparatus. We found that mouse behaviors on these apparatuses were surprisingly different. It took a markedly longer time to train mice on the narrow- than wide-compartment rotarod. Further, non-transgenic B6129S and tau knockout mice aged 11 months and beyond showed different levels of improvement based on the compartment width. These mice had no overt improvements on accelerating rotarod over 4-5 training sessions on the narrow compartment, contrary to marked progress on the wide counterpart. The incidents of mice passively somersaulting round and fragmented running occurred significantly more on the wide than narrow compartment during accelerating rotarod sessions. Mice fell off rod more frequently on narrow than wide compartments upon attempt to turn around and when moving backward on rod. The pros and cons of narrow versus wide compartments are informative as to how to choose a rotarod apparatus that best fits the animal models used.
Collapse
Affiliation(s)
- Sachiko P Keane
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Kathryn K Chadman
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Andres R Gomez
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wen Hu
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA.
| |
Collapse
|
11
|
Chen BH, Lin ZY, Zeng XX, Jiang YH, Geng F. LRP4-related signalling pathways and their regulatory role in neurological diseases. Brain Res 2024; 1825:148705. [PMID: 38065285 DOI: 10.1016/j.brainres.2023.148705] [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: 10/04/2023] [Revised: 11/17/2023] [Accepted: 12/03/2023] [Indexed: 01/28/2024]
Abstract
The mechanism of action of low-density lipoprotein receptor related protein 4 (LRP4) is mediated largely via the Agrin-LRP4-MuSK signalling pathway in the nervous system. LRP4 contributes to the development of synapses in the peripheral nervous system (PNS). It interacts with signalling molecules such as the amyloid beta-protein precursor (APP) and the wingless type protein (Wnt). Its mechanisms of action are complex and mediated via interaction between the pre-synaptic motor neuron and post-synaptic muscle cell in the PNS, which enhances the development of the neuromuscular junction (NMJ). LRP4 may function differently in the central nervous system (CNS) than in the PNS, where it regulates ATP and glutamate release via astrocytes. It mayaffect the growth and development of the CNS by controlling the energy metabolism. LRP4 interacts with Agrin to maintain dendrite growth and density in the CNS. The goal of this article is to review the current studies involving relevant LRP4 signaling pathways in the nervous system. The review also discusses the clinical and etiological roles of LRP4 in neurological illnesses, such as myasthenia gravis, Alzheimer's disease and epilepsy. In this review, we provide a theoretical foundation for the pathogenesis and therapeutic application of LRP4 in neurologic diseases.
Collapse
Affiliation(s)
- Bai-Hui Chen
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Ze-Yu Lin
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Xiao-Xue Zeng
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Yi-Han Jiang
- Department of Physiology, Shantou University Medical College, Shantou 515041, China
| | - Fei Geng
- Department of Physiology, Shantou University Medical College, Shantou 515041, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
12
|
Cossins J, Kozma I, Canzonetta C, Hawkins A, Beeson D, Sepulveda P, Dong Y. Dose escalation pre-clinical trial of novel DOK7-AAV in mouse model of DOK7 congenital myasthenia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579626. [PMID: 38405691 PMCID: PMC10888934 DOI: 10.1101/2024.02.09.579626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Congenital myasthenic syndromes (CMS) are a group of inherited disorders characterised by defective neuromuscular transmission and fatigable muscle weakness. Mutations in DOK7 , a gene encoding a post-synaptic protein crucial in the formation and stabilisation of the neuromuscular junction (NMJ), rank among the leading three prevalent causes of CMS in diverse populations globally. The majority of DOK7 CMS patients experience varying degrees of disability despite receiving optimised treatment, necessitating the development of improved therapeutic approaches. Here we executed a dose escalation pre-clinical trial using a DOK7-CMS mouse model to assess the efficacy of Amp-101, an innovative AAV gene replacement therapy. Amp-101 is based on AAVrh74 and contains human DOK7 cDNA under the control of a muscle-restricted promoter. We show that at doses 6x10 13 vg/kg and 1x10 14 vg/kg, Amp-101 generated enlarged NMJs and rescued the very severe phenotype of the model. Treated mice became at least as strong as WT littermates and the diaphragm and tibialis anterior muscles displayed robust expression of DOK7. This data suggests that Amp-101 is a promising candidate to move forward to clinic trials.
Collapse
|
13
|
Zhang Y, Lu Y, Yu M, Wang J, Du X, Zhao D, Pian H, He Z, Wu G, Li S, Wang S, Yu D. Transcriptome Profiling Identifies Differentially Expressed Genes in Skeletal Muscle Development in Native Chinese Ducks. Genes (Basel) 2023; 15:52. [PMID: 38254942 PMCID: PMC10815232 DOI: 10.3390/genes15010052] [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: 11/02/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
China boasts a rich diversity of indigenous duck species, some of which exhibit desirable economic traits. Here, we generated transcriptome sequencing datasets of breast muscle tissue samples from 1D of four groups: Pekin duck pure breeding group (P), Jinling White duck breeding group (J), P ♂ × J ♀ orthogonal group (PJ) and J ♂ × P ♀ reciprocal-cross group (JP) (n = 3), chosen based on the distinctive characteristics of duck muscle development during the embryonic period. We identified 5053 differentially expressed genes (DEGs) among the four groups. Network prediction analysis showed that ribosome and oxidative phosphorylation-related genes were the most enriched, and muscular protein-related genes were found in the 14-day-old embryonic group. We found that previously characterized functional genes, such as FN1, AGRN, ADNAMST3, APOB and FGF9, were potentially involved in muscle development in 14-day-old embryos. Functional enrichment analysis suggested that genes that participated in molecular function and cell component and key signaling pathways (e.g., hippo, ribosome, oxidative phosphorylation) were significantly enriched in the development of skeletal muscle at 14 days of embryonic age. These results indicate a possible role of muscle metabolism and myoglobin synthesis in skeletal muscle development in both duck parents and hybrids.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Yinglin Lu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Minli Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Jin Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Xubin Du
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Dong Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
- School of Animal Medical, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China
| | - Huifang Pian
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| | - Zongliang He
- Nanjing Academy of Animal Husbandry and Poultry, Nanjing 210095, China
| | - Guansuo Wu
- Nanjing Academy of Animal Husbandry and Poultry, Nanjing 210095, China
| | - Shiwei Li
- College of Animal Science, Xizang Agricultural and Animal Husbandry University, Linzhi 860000, China
| | - Sike Wang
- College of Animal Science, Xizang Agricultural and Animal Husbandry University, Linzhi 860000, China
| | - Debing Yu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.Z.)
| |
Collapse
|
14
|
Ravn-Boess N, Roy N, Hattori T, Bready D, Donaldson H, Lawson C, Lapierre C, Korman A, Rodrick T, Liu E, Frenster JD, Stephan G, Wilcox J, Corrado AD, Cai J, Ronnen R, Wang S, Haddock S, Sabio Ortiz J, Mishkit O, Khodadadi-Jamayran A, Tsirigos A, Fenyö D, Zagzag D, Drube J, Hoffmann C, Perna F, Jones DR, Possemato R, Koide A, Koide S, Park CY, Placantonakis DG. The expression profile and tumorigenic mechanisms of CD97 (ADGRE5) in glioblastoma render it a targetable vulnerability. Cell Rep 2023; 42:113374. [PMID: 37938973 PMCID: PMC10841603 DOI: 10.1016/j.celrep.2023.113374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/08/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain malignancy. Adhesion G protein-coupled receptors (aGPCRs) have attracted interest for their potential as treatment targets. Here, we show that CD97 (ADGRE5) is the most promising aGPCR target in GBM, by virtue of its de novo expression compared to healthy brain tissue. CD97 knockdown or knockout significantly reduces the tumor initiation capacity of patient-derived GBM cultures (PDGCs) in vitro and in vivo. We find that CD97 promotes glycolytic metabolism via the mitogen-activated protein kinase (MAPK) pathway, which depends on phosphorylation of its C terminus and recruitment of β-arrestin. We also demonstrate that THY1/CD90 is a likely CD97 ligand in GBM. Lastly, we show that an anti-CD97 antibody-drug conjugate selectively kills tumor cells in vitro. Our studies identify CD97 as a regulator of tumor metabolism, elucidate mechanisms of receptor activation and signaling, and provide strong scientific rationale for developing biologics to target it therapeutically in GBM.
Collapse
Affiliation(s)
- Niklas Ravn-Boess
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Nainita Roy
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Takamitsu Hattori
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Devin Bready
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Hayley Donaldson
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Christopher Lawson
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Cathryn Lapierre
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Aryeh Korman
- Metabolomics Laboratory, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Tori Rodrick
- Metabolomics Laboratory, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Enze Liu
- Department of Medicine, Division of Hematology/Oncology, Indiana University, Indianapolis, IN 46202, USA
| | - Joshua D Frenster
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Gabriele Stephan
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jordan Wilcox
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Alexis D Corrado
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Julia Cai
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Rebecca Ronnen
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Shuai Wang
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Sara Haddock
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jonathan Sabio Ortiz
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Orin Mishkit
- Preclinical Imaging Laboratory, NYU Grossman School of Medicine, New York, NY 10016, USA
| | | | - Aris Tsirigos
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA; Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - David Zagzag
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Julia Drube
- Institute for Molecular Cell Biology, Universitätsklinikum Jena, 07745 Jena, Germany
| | - Carsten Hoffmann
- Institute for Molecular Cell Biology, Universitätsklinikum Jena, 07745 Jena, Germany
| | | | - Drew R Jones
- Metabolomics Laboratory, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Richard Possemato
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Akiko Koide
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Shohei Koide
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Christopher Y Park
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Dimitris G Placantonakis
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY 10016, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Kimmel Center for Stem Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA; Brain and Spine Tumor Center, NYU Grossman School of Medicine, New York, NY 10016, USA; Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
15
|
Qaisar R. Targeting neuromuscular junction to treat neuromuscular disorders. Life Sci 2023; 333:122186. [PMID: 37858716 DOI: 10.1016/j.lfs.2023.122186] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
The integrity and preservation of the neuromuscular junction (NMJ), the interface between the motor neuron and skeletal muscle, is critical for maintaining a healthy skeletal muscle. The structural and/or functional defects in the three cellular components of NMJ, namely the pre-synaptic terminal, synaptic cleft, and post-synaptic region, negatively affect skeletal muscle mass and/or strength. Therefore, NMJ repair appears to be an appropriate therapy for muscle disorders. Mouse models provide a detailed molecular characterization of various cellular components of NMJ with relevance to human diseases. This review discusses different molecular targets on the three cellular components of NMJ for treating muscle diseases. The potential effects of these therapies on NMJ morphology and motor performance, their therapeutic efficacy, and clinical relevance are discussed. Collectively, the available data supports targeting NMJ alone or as an adjunct therapy in treating muscle disorders. However, the potential impact of such interventions on human patients with muscle disorders requires further investigation.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Space Medicine Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Cardiovascular Research Group, Sharjah Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
16
|
Bakooshli MA, Wang YX, Monti E, Su S, Kraft P, Nalbandian M, Alexandrova L, Wheeler JR, Vogel H, Blau HM. Regeneration of neuromuscular synapses after acute and chronic denervation by inhibiting the gerozyme 15-prostaglandin dehydrogenase. Sci Transl Med 2023; 15:eadg1485. [PMID: 37820010 PMCID: PMC10763629 DOI: 10.1126/scitranslmed.adg1485] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 09/22/2023] [Indexed: 10/13/2023]
Abstract
To date, there are no approved treatments for the diminished strength and paralysis that result from the loss of peripheral nerve function due to trauma, heritable neuromuscular diseases, or aging. Here, we showed that denervation resulting from transection of the sciatic nerve triggered a marked increase in the prostaglandin-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in skeletal muscle in mice, providing evidence that injury drives early expression of this aging-associated enzyme or gerozyme. Treating mice with a small-molecule inhibitor of 15-PGDH promoted regeneration of motor axons and formation of neuromuscular synapses leading to an acceleration in recovery of force after an acute nerve crush injury. In aged mice with chronic denervation of muscles, treatment with the 15-PGDH inhibitor increased motor neuron viability and restored neuromuscular junctions and function. These presynaptic changes synergized with previously reported muscle tissue remodeling to result in a marked increase in the strength of aged muscles. We further found that 15-PGDH aggregates defined the target fibers that are histopathologic hallmarks of human neurogenic myopathies, suggesting that the gerozyme may be involved in their etiology. Our data suggest that inhibition of 15-PGDH may constitute a therapeutic strategy to physiologically boost prostaglandin E2, restore neuromuscular connectivity, and promote recovery of strength after acute or chronic denervation due to injury, disease, or aging.
Collapse
Affiliation(s)
- Mohsen A. Bakooshli
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yu Xin Wang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Elena Monti
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shiqi Su
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peggy Kraft
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Minas Nalbandian
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ludmila Alexandrova
- Vincent Coates Foundation Mass Spectrometry Laboratory, Stanford University, Stanford, CA 94305, USA
| | - Joshua R. Wheeler
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
- Department of Neuropathology, Stanford University, Stanford, CA 94305, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
- Department of Neuropathology, Stanford University, Stanford, CA 94305, USA
| | - Helen M. Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
17
|
Huang YT, Crick HR, Chaytow H, van der Hoorn D, Alhindi A, Jones RA, Hector RD, Cobb SR, Gillingwater TH. Long-term muscle-specific overexpression of DOK7 in mice using AAV9-tMCK-DOK7. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:617-628. [PMID: 37637210 PMCID: PMC10457688 DOI: 10.1016/j.omtn.2023.07.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023]
Abstract
Neuromuscular junction (NMJ) dysfunction underlies several diseases, including congenital myasthenic syndromes (CMSs) and motor neuron disease (MND). Molecular pathways governing NMJ stability are therefore of interest from both biological and therapeutic perspectives. Muscle-specific kinase (MuSK) is necessary for the formation and maintenance of post-synaptic elements of the NMJ, and downstream of tyrosine kinases 7 (DOK7) is crucial for activation of the MuSK pathway. Overexpression of DOK7 using AAV9 has been shown to ameliorate neuromuscular pathology in pre-clinical disease models of CMS and MND. However, long-term consequences of DOK7 expression have been sparsely investigated and targeted overexpression of DOK7 in skeletal muscle yet to be established. Here, we developed and characterized a novel AAV9-DOK7 facilitating forced expression of DOK7 under a skeletal muscle-specific promoter. AAV9-tMCK-DOK7 was systemically delivered to newborn mice that were monitored over 6 months. DOK7 overexpression was restricted to skeletal muscles. Body weight, blood biochemistry, and histopathological assessments were unaffected by AAV9-tMCK-DOK7 treatment. In contrast, forced expression of DOK7 resulted in enlargement of both the pre- and post-synaptic components of the NMJ, without causing denervation. We conclude that muscle-specific DOK7 overexpression can be achieved in a safe manner, with the capacity to target NMJs in vivo.
Collapse
Affiliation(s)
- Yu-Ting Huang
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Hannah R. Crick
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Helena Chaytow
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Dinja van der Hoorn
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Abrar Alhindi
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
- Faculty of Medicine, Department of Anatomy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ross A. Jones
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | | | | | - Thomas H. Gillingwater
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| |
Collapse
|
18
|
Spendiff S, Dong Y, Maggi L, Rodríguez Cruz PM, Beeson D, Lochmüller H. 260th ENMC International Workshop: Congenital myasthenic syndromes 11-13 March 2022, Hoofddorp, The Netherlands. Neuromuscul Disord 2023; 33:111-118. [PMID: 36609117 DOI: 10.1016/j.nmd.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Sally Spendiff
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Yin Dong
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Pedro M Rodríguez Cruz
- Centro Nacional de Análisis Genómico (CNAG-CRG), Centre for Genomic Regulation, Barcelona, Spain; Department of Human Genetics, Université Cheikh Anta Diop, Dakar, Senegal; Department of Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada; Department of Medicine, Division of Neurology, The Ottawa Hospital, Ottawa, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada; Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.
| |
Collapse
|
19
|
Xie T, Xu G, Liu Y, Quade B, Lin W, Bai XC. Structural insights into the assembly of the agrin/LRP4/MuSK signaling complex. Proc Natl Acad Sci U S A 2023; 120:e2300453120. [PMID: 37252960 PMCID: PMC10266037 DOI: 10.1073/pnas.2300453120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
MuSK is a receptor tyrosine kinase (RTK) that plays essential roles in the formation and maintenance of the neuromuscular junction. Distinct from most members of RTK family, MuSK activation requires not only its cognate ligand agrin but also its coreceptors LRP4. However, how agrin and LRP4 coactivate MuSK remains unclear. Here, we report the cryo-EM structure of the extracellular ternary complex of agrin/LRP4/MuSK in a stoichiometry of 1:1:1. This structure reveals that arc-shaped LRP4 simultaneously recruits both agrin and MuSK to its central cavity, thereby promoting a direct interaction between agrin and MuSK. Our cryo-EM analyses therefore uncover the assembly mechanism of agrin/LRP4/MuSK signaling complex and reveal how MuSK receptor is activated by concurrent binding of agrin and LRP4.
Collapse
Affiliation(s)
- Tian Xie
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Guangjun Xu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Yun Liu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Bradley Quade
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Weichun Lin
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Xiao-chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
20
|
Lim JL, Augustinus R, Plomp JJ, Roya-Kouchaki K, Vergoossen DLE, Fillié-Grijpma Y, Struijk J, Thomas R, Salvatori D, Steyaert C, Blanchetot C, Vanhauwaert R, Silence K, van der Maarel SM, Verschuuren JJ, Huijbers MG. Development and characterization of agonistic antibodies targeting the Ig-like 1 domain of MuSK. Sci Rep 2023; 13:7478. [PMID: 37156800 PMCID: PMC10167245 DOI: 10.1038/s41598-023-32641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/30/2023] [Indexed: 05/10/2023] Open
Abstract
Muscle-specific kinase (MuSK) is crucial for acetylcholine receptor (AChR) clustering and thereby neuromuscular junction (NMJ) function. NMJ dysfunction is a hallmark of several neuromuscular diseases, including MuSK myasthenia gravis. Aiming to restore NMJ function, we generated several agonist monoclonal antibodies targeting the MuSK Ig-like 1 domain. These activated MuSK and induced AChR clustering in cultured myotubes. The most potent agonists partially rescued myasthenic effects of MuSK myasthenia gravis patient IgG autoantibodies in vitro. In an IgG4 passive transfer MuSK myasthenia model in NOD/SCID mice, MuSK agonists caused accelerated weight loss and no rescue of myasthenic features. The MuSK Ig-like 1 domain agonists unexpectedly caused sudden death in a large proportion of male C57BL/6 mice (but not female or NOD/SCID mice), likely caused by a urologic syndrome. In conclusion, these agonists rescued pathogenic effects in myasthenia models in vitro, but not in vivo. The sudden death in male mice of one of the tested mouse strains revealed an unexpected and unexplained role for MuSK outside skeletal muscle, thereby hampering further (pre-) clinical development of these clones. Future research should investigate whether other Ig-like 1 domain MuSK antibodies, binding different epitopes, do hold a safe therapeutic promise.
Collapse
Affiliation(s)
- Jamie L Lim
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Roy Augustinus
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Jaap J Plomp
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kasra Roya-Kouchaki
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Dana L E Vergoossen
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Yvonne Fillié-Grijpma
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Josephine Struijk
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Rachel Thomas
- Department PDC-Pathologie, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniela Salvatori
- Veterinary Faculty, Department Clinical Sciences, Universiteit Utrecht, Utrecht, The Netherlands
| | | | | | | | | | - Silvère M van der Maarel
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Jan J Verschuuren
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands.
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
21
|
Muñoz-García MI, Guerrero-Molina MP, de Fuenmayor-Fernández de la Hoz CP, Bermejo-Guerrero L, Arteche-López A, Hernández-Laín A, Martín MA, Domínguez-González C. Delayed Diagnosis of Congenital Myasthenic Syndromes Erroneously Interpreted as Mitochondrial Myopathies. J Clin Med 2023; 12:jcm12093308. [PMID: 37176748 PMCID: PMC10179722 DOI: 10.3390/jcm12093308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/02/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Congenital myasthenic syndromes (CMSs) and primary mitochondrial myopathies (PMMs) can present with ptosis, external ophthalmoplegia, and limb weakness. METHODS Our method involved the description of three cases of CMS that were initially characterized as probable PMM. RESULTS All patients were male and presented with ptosis and/or external ophthalmoplegia at birth, with proximal muscle weakness and fatigue on physical exertion. After normal repetitive nerve stimulation (RNS) studies performed on facial muscles, a muscle biopsy (at a median age of 9) was performed to rule out congenital myopathies. In all three cases, the biopsy findings (COX-negative fibers or respiratory chain defects) pointed to PMM. They were referred to our neuromuscular unit in adulthood to establish a genetic diagnosis. However, at this time, fatigability was evident in the physical exams and RNS in the spinal accessory nerve showed a decremental response in all cases. Targeted genetic studies revealed pathogenic variants in the MUSK, DOK7, and RAPSN genes. The median diagnostic delay was 29 years. Treatment resulted in functional improvement in all cases. CONCLUSIONS Early identification of CMS is essential as medical treatment can provide clear benefits. Its diagnosis can be challenging due to phenotypic overlap with other debilitating disorders. Thus, a high index of suspicion is necessary to guide the diagnostic strategy.
Collapse
Affiliation(s)
- Mariana I Muñoz-García
- Neuromuscular Unit, Neurology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | | | | | - Laura Bermejo-Guerrero
- Neuromuscular Unit, Neurology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Ana Arteche-López
- Genetics Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | | | - Miguel A Martín
- Genetics Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), 28041 Madrid, Spain
- Mitochondrial and Neuromuscular Diseases Research Group, 12 de Octubre, Hospital Research Institute (imas12), 28041 Madrid, Spain
| | - Cristina Domínguez-González
- Neuromuscular Unit, Neurology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), 28041 Madrid, Spain
- Mitochondrial and Neuromuscular Diseases Research Group, 12 de Octubre, Hospital Research Institute (imas12), 28041 Madrid, Spain
| |
Collapse
|
22
|
Mendpara V, Bethanabotla S, Yadav M, Kanisetti V, Singh G, Das A, Sahu S, Patel H. When Breathing Becomes a Challenge: A Case of Congenital Myasthenia Gravis in an Indian Neonate With a DOK-7 Gene Mutation. Cureus 2023; 15:e38842. [PMID: 37303354 PMCID: PMC10256249 DOI: 10.7759/cureus.38842] [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] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
A rare neuromuscular condition known as congenital myasthenia gravis (CMG) affects some people from birth or very soon after. It results in fatigue and muscle weakness because of genetic abnormalities that interfere with the neuromuscular junction's ability to function, where the nerves and muscles connect. Even among those who have the same genetic mutation, the severity of CMG symptoms might differ considerably. The most typical signs of CMG include eyelid drooping, breathing issues, muscle weakness and weariness, and difficulties swallowing. Clinical examinations, neurophysiologic tests, and genetic analyses are frequently combined to make the diagnosis of CMG. Although there is no known treatment for CMG, many patients may control their symptoms and lead relatively normal lives with the right care. A newborn with CMG due to a DOK-7 gene mutation is described in this article, along with its very early onset. The DOK-7 mutation is a rare variant in the Indian population that causes CMG and usually manifests as 'limb girdle' weakness. However, due to muscle weakness, the neonate in this case developed severe respiratory distress and later died despite rigorous life-saving measures.
Collapse
Affiliation(s)
- Vaidehi Mendpara
- Medicine and Surgery, Pediatrics, Government Medical College Surat, Surat, IND
| | | | - Megha Yadav
- Medicine and Surgery, Maharani Laxmi Bai Medical College, Jhansi, IND
| | | | - Gurpreet Singh
- Medicine, Government Medical College and Hospital, Amritsar, IND
| | - Abhirami Das
- Internal Medicine, Rajiv Gandhi Medical College, Thane, IND
| | - Sweta Sahu
- Surgery, Jagadguru Jayadeva Murugarajendra (JJM) Medical College, Davanagere, IND
| | - Hitesh Patel
- Pediatrics and Child Health, Government Medical College and Hospital, Surat, IND
| |
Collapse
|
23
|
Prömer J, Barresi C, Herbst R. From phosphorylation to phenotype - Recent key findings on kinase regulation, downstream signaling and disease surrounding the receptor tyrosine kinase MuSK. Cell Signal 2023; 104:110584. [PMID: 36608736 DOI: 10.1016/j.cellsig.2022.110584] [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: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/31/2022] [Indexed: 01/04/2023]
Abstract
Muscle-specific kinase (MuSK) is the key regulator of neuromuscular junction development. MuSK acts via several distinct pathways and is responsible for pre- and postsynaptic differentiation. MuSK is unique among receptor tyrosine kinases as activation and signaling are particularly tightly regulated. Initiation of kinase activity requires Agrin, a heparan sulphate proteoglycan derived from motor neurons, the low-density lipoprotein receptor-related protein-4 (Lrp4) and the intracellular adaptor protein Dok-7. There is a great knowledge gap between MuSK activation and downstream signaling. Recent studies using omics techniques have addressed this knowledge gap, thereby greatly contributing to a better understanding of MuSK signaling. Impaired MuSK signaling causes severe muscle weakness as described in congenital myasthenic syndromes or myasthenia gravis but the underlying pathophysiology is often unclear. This review focuses on recent advances in deciphering MuSK activation and downstream signaling. We further highlight latest break-throughs in understanding and treatment of MuSK-related disorders and discuss the role of MuSK in non-muscle tissue.
Collapse
Affiliation(s)
- Jakob Prömer
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Cinzia Barresi
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ruth Herbst
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Mori S, Suzuki S, Konishi T, Kawaguchi N, Kishi M, Kuwabara S, Ishizuchi K, Zhou H, Shibasaki F, Tsumoto H, Omura T, Miura Y, Mori S, Higashihara M, Murayama S, Shigemoto K. Proteolytic ectodomain shedding of muscle-specific tyrosine kinase in myasthenia gravis. Exp Neurol 2023; 361:114300. [PMID: 36525997 DOI: 10.1016/j.expneurol.2022.114300] [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: 07/12/2022] [Revised: 11/07/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
Autoantibodies to muscle-specific tyrosine kinase (MuSK) proteins at the neuromuscular junction (NMJ) cause refractory generalized myasthenia gravis (MG) with dyspnea more frequently than other MG subtypes. However, the mechanisms via which MuSK, a membrane protein locally expressed on the NMJ of skeletal muscle, is supplied to the immune system as an autoantigen remains unknown. Here, we identified MuSK in both mouse and human serum, with the amount of MuSK dramatically increasing in mice with motor nerve denervation and in MG model mice. Peptide analysis by liquid chromatography-tandem-mass spectrometry (LC-MS/MS) confirmed the presence of MuSK in both human and mouse serum. Furthermore, some patients with MG have significantly higher amounts of MuSK in serum than healthy controls. Our results indicated that the secretion of MuSK proteins from muscles into the bloodstream was induced by ectodomain shedding triggered by neuromuscular junction failure. The results may explain why MuSK-MG is refractory to treatments and causes rapid muscle atrophy in some patients due to the denervation associated with Ab-induced disruption of neuromuscular transmission at the NMJ. Such discoveries pave the way for new MG treatments, and MuSK may be used as a biomarker for other neuromuscular diseases in preclinical studies, clinical diagnostics, therapeutics, and drug discovery.
Collapse
Affiliation(s)
- Shuuichi Mori
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Shigeaki Suzuki
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | | | - Naoki Kawaguchi
- Dowa Institute of Clinical Neuroscience, Neurology Clinic Chiba, Chiba, Japan
| | - Masahiko Kishi
- Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Chiba University School of Medicine, Chiba, Japan
| | - Kei Ishizuchi
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Heying Zhou
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Futoshi Shibasaki
- Department of Genome Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiroki Tsumoto
- Research Team for Mechanism of Aging, TMIG, Tokyo, Japan
| | - Takuya Omura
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, TMIG, Tokyo, Japan
| | - Seijiro Mori
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Geriatric Hospital, Tokyo (TMGHIG), Japan
| | | | - Kazuhiro Shigemoto
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan.
| |
Collapse
|
25
|
Ohno K, Ohkawara B, Shen XM, Selcen D, Engel AG. Clinical and Pathologic Features of Congenital Myasthenic Syndromes Caused by 35 Genes-A Comprehensive Review. Int J Mol Sci 2023; 24:ijms24043730. [PMID: 36835142 PMCID: PMC9961056 DOI: 10.3390/ijms24043730] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Congenital myasthenic syndromes (CMS) are a heterogeneous group of disorders characterized by impaired neuromuscular signal transmission due to germline pathogenic variants in genes expressed at the neuromuscular junction (NMJ). A total of 35 genes have been reported in CMS (AGRN, ALG14, ALG2, CHAT, CHD8, CHRNA1, CHRNB1, CHRND, CHRNE, CHRNG, COL13A1, COLQ, DOK7, DPAGT1, GFPT1, GMPPB, LAMA5, LAMB2, LRP4, MUSK, MYO9A, PLEC, PREPL, PURA, RAPSN, RPH3A, SCN4A, SLC18A3, SLC25A1, SLC5A7, SNAP25, SYT2, TOR1AIP1, UNC13A, VAMP1). The 35 genes can be classified into 14 groups according to the pathomechanical, clinical, and therapeutic features of CMS patients. Measurement of compound muscle action potentials elicited by repetitive nerve stimulation is required to diagnose CMS. Clinical and electrophysiological features are not sufficient to identify a defective molecule, and genetic studies are always required for accurate diagnosis. From a pharmacological point of view, cholinesterase inhibitors are effective in most groups of CMS, but are contraindicated in some groups of CMS. Similarly, ephedrine, salbutamol (albuterol), amifampridine are effective in most but not all groups of CMS. This review extensively covers pathomechanical and clinical features of CMS by citing 442 relevant articles.
Collapse
Affiliation(s)
- Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Correspondence: (K.O.); (A.G.E.)
| | - Bisei Ohkawara
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Xin-Ming Shen
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | - Duygu Selcen
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | - Andrew G. Engel
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: (K.O.); (A.G.E.)
| |
Collapse
|
26
|
Cao R, Chen P, Wang H, Jing H, Zhang H, Xing G, Luo B, Pan J, Yu Z, Xiong WC, Mei L. Intrafusal-fiber LRP4 for muscle spindle formation and maintenance in adult and aged animals. Nat Commun 2023; 14:744. [PMID: 36765071 PMCID: PMC9918736 DOI: 10.1038/s41467-023-36454-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Proprioception is sensed by muscle spindles for precise locomotion and body posture. Unlike the neuromuscular junction (NMJ) for muscle contraction which has been well studied, mechanisms of spindle formation are not well understood. Here we show that sensory nerve terminals are disrupted by the mutation of Lrp4, a gene required for NMJ formation; inducible knockout of Lrp4 in adult mice impairs sensory synapses and movement coordination, suggesting that LRP4 is required for spindle formation and maintenance. LRP4 is critical to the expression of Egr3 during development; in adult mice, it interacts in trans with APP and APLP2 on sensory terminals. Finally, spindle sensory endings and function are impaired in aged mice, deficits that could be diminished by LRP4 expression. These observations uncovered LRP4 as an unexpected regulator of muscle spindle formation and maintenance in adult and aged animals and shed light on potential pathological mechanisms of abnormal muscle proprioception.
Collapse
Affiliation(s)
- Rangjuan Cao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peng Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongyang Jing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongsheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Guanglin Xing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jinxiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Zheng Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
27
|
Wang W, Miyamoto Y, Chen B, Shi J, Diao F, Zheng W, Li Q, Yu L, Li L, Xu Y, Wu L, Mao X, Fu J, Li B, Yan Z, Shi R, Xue X, Mu J, Zhang Z, Wu T, Zhao L, Wang W, Zhou Z, Dong J, Li Q, Jin L, He L, Sun X, Lin G, Kuang Y, Wang L, Sang Q. Karyopherin α deficiency contributes to human preimplantation embryo arrest. J Clin Invest 2023; 133:159951. [PMID: 36647821 PMCID: PMC9843055 DOI: 10.1172/jci159951] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 11/02/2022] [Indexed: 01/18/2023] Open
Abstract
Preimplantation embryo arrest (PREMBA) is a common cause of female infertility and recurrent failure of assisted reproductive technology. However, the genetic basis of PREMBA is largely unrevealed. Here, using whole-exome sequencing data from 606 women experiencing PREMBA compared with 2,813 controls, we performed a population and gene-based burden test and identified a candidate gene, karyopherin subunit α7 (KPNA7). In vitro studies showed that identified sequence variants reduced KPNA7 protein levels, impaired KPNA7 capacity for binding to its substrate ribosomal L1 domain-containing protein 1 (RSL1D1), and affected KPNA7 nuclear transport activity. Comparison between humans and mice suggested that mouse KPNA2, rather than mouse KPNA7, acts as an essential karyopherin in embryonic development. Kpna2-/- female mice showed embryo arrest due to zygotic genome activation defects, recapitulating the phenotype of human PREMBA. In addition, female mice with an oocyte-specific knockout of Rsl1d1 recapitulated the phenotype of Kpna2-/- mice, demonstrating the vital role of substrate RSL1D1. Finally, complementary RNA (cRNA) microinjection of human KPNA7, but not mouse Kpna7, was able to rescue the embryo arrest phenotype in Kpna2-/- mice, suggesting mouse KPNA2 might be a homologue of human KPNA7. Our findings uncovered a mechanistic understanding for the pathogenesis of PREMBA, which acts by impairing nuclear protein transport, and provide a diagnostic marker for PREMBA patients.
Collapse
Affiliation(s)
- Wenjing Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Yoichi Miyamoto
- Laboratory of Nuclear Transport Dynamics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Juanzi Shi
- Reproductive Medicine Center, Northwest Women’s and Children’s Hospital, Xi’an, China
| | - Feiyang Diao
- Reproductive Medicine Center, Jiangsu Province Hospital, Jiangsu, China
| | - Wei Zheng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Qun Li
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Lan Yu
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lin Li
- Key Laboratory of Human Reproduction and Genetics, Department of Reproductive Medicine, Nanchang Reproductive Hospital, Nanchang, China
| | - Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ling Wu
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyan Mao
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Fu
- Shanghai Ji’ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, and
| | - Bin Li
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Yan
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Shi
- Reproductive Medicine Center, Northwest Women’s and Children’s Hospital, Xi’an, China
| | - Xia Xue
- Reproductive Medicine Center, Northwest Women’s and Children’s Hospital, Xi’an, China
| | - Jian Mu
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Zhihua Zhang
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Tianyu Wu
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Lin Zhao
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Weijie Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Zhou Zhou
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Jie Dong
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Qiaoli Li
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxi Sun
- Shanghai Ji’ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, and
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yanping Kuang
- Reproductive Medicine Center, Shanghai Ninth Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai, China
| |
Collapse
|
28
|
De novo Fc-based receptor dimerizers differentially modulate PlexinB1 function. Structure 2022; 30:1411-1423.e4. [PMID: 35981535 DOI: 10.1016/j.str.2022.07.008] [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: 10/14/2021] [Revised: 06/26/2022] [Accepted: 07/22/2022] [Indexed: 11/21/2022]
Abstract
Signaling by single-pass transmembrane receptors often involves a formation of ligand-induced receptor dimers with particular conformation, and bivalent receptor binders can modulate receptor functions by inducing different receptor dimer conformations, although such agents are difficult to design. Here, we describe the generation of both antagonistic and agonistic receptor dimerizers toward PlexinB1 (PlxnB1), a receptor for semaphorin 4D (Sema4D), by grafting two different PlxnB1-binding peptides onto the human immunoglobulin G1 (IgG1) Fc protein. The function-modulating activity of a peptide Fc was strongly dependent on the type of the peptide as well as the grafting site, with the best variants showing activity at an nM concentration range. Structural analysis of each peptide-PlxnB1 complex revealed that the agonistic Fc dimerizes PlxnB1 in a face-to-face fashion similar to that induced by Sema4D, whereas antagonistic Fc would induce signaling-incompetent PlxnB1 dimer conformation, enforcing the idea that plexin activation is primarily controlled by the receptor orientation within the dimer.
Collapse
|
29
|
Control of CRK-RAC1 activity by the miR-1/206/133 miRNA family is essential for neuromuscular junction function. Nat Commun 2022; 13:3180. [PMID: 35676269 PMCID: PMC9178026 DOI: 10.1038/s41467-022-30778-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 05/07/2022] [Indexed: 11/08/2022] Open
Abstract
Formation and maintenance of neuromuscular junctions (NMJs) are essential for skeletal muscle function, allowing voluntary movements and maintenance of the muscle tone, thereby preventing atrophy. Generation of NMJs depends on the interaction of motor neurons with skeletal muscle fibers, which initiates a cascade of regulatory events that is essential for patterning of acetylcholine receptor (AChR) clusters at specific sites of the sarcolemma. Here, we show that muscle-specific miRNAs of the miR-1/206/133 family are crucial regulators of a signaling cascade comprising DOK7-CRK-RAC1, which is critical for stabilization and anchoring of postsynaptic AChRs during NMJ development and maintenance. We describe that posttranscriptional repression of CRK by miR-1/206/133 is essential for balanced activation of RAC1. Failure to adjust RAC1 activity severely compromises NMJ function, causing respiratory failure in neonates and neuromuscular symptoms in adult mice. We conclude that miR-1/206/133 serve a specific function for NMJs but are dispensable for skeletal muscle development. The miR-1/133/206 gene family codes for the most abundant microRNAs in striated muscles. Here, Klockner et al show that inactivation of all family members in skeletal muscle prevents formation of normal neuromuscular junctions due to increased expression of the adaptor protein CRK.
Collapse
|
30
|
Tayade K, Salunkhe M, Agarwal A, Radhakrishnan DM, Srivastava AK. DOK7 congenital myasthenic syndrome responsive to oral salbutamol. QJM 2022; 115:323-324. [PMID: 35092298 DOI: 10.1093/qjmed/hcac017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- K Tayade
- From the Department of Neurology, Cardioneurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - M Salunkhe
- From the Department of Neurology, Cardioneurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - A Agarwal
- From the Department of Neurology, Cardioneurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - D M Radhakrishnan
- From the Department of Neurology, Cardioneurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - A K Srivastava
- From the Department of Neurology, Cardioneurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
31
|
Sfeir A, Fishell G, Schier AF, Dustin ML, Gan WB, Joyner A, Lehmann R, Ron D, Roth D, Talbot WS, Yelon D, Zychlinsky A. Basic science under threat: Lessons from the Skirball Institute. Cell 2022; 185:755-758. [DOI: 10.1016/j.cell.2022.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 11/28/2022]
|
32
|
Ramdas S, Beeson D. Congenital myasthenic syndromes: where do we go from here? Neuromuscul Disord 2021; 31:943-954. [PMID: 34736634 DOI: 10.1016/j.nmd.2021.07.400] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 11/27/2022]
Abstract
Congenital myasthenia syndromes are rare but often treatable conditions affecting neuromuscular transmission. They result from loss or impaired function of one of a number of proteins secondary to a genetic defect. An estimate of the prevalence in the UK gave 9.2 cases per million, however, this is likely an underestimate since the adoption of next generation sequencing for diagnosis away from specialist centres is enhancing the 'pick up' rate. Next generation sequencing has helped identify a series of novel genes that harbour mutations causative for congenital myasthenic syndrome that include not only genes that encode proteins specifically expressed at the neuromuscular junction but also those that are ubiquitously expressed. The list of genes harbouring disease-causing mutations for congenital myasthenic syndrome continues to expand and is now over 30, but with many of the newly identified genes it is increasingly being recognised that abnormal neuromuscular transmission is only one component of a multifaceted phenotype in which muscle, the central nervous system, and other organs may also be affected. Treatment can be tailored to the underlying molecular mechanism for impaired neuromuscular transmission but treating the more complex multifaceted disorders and will require development of new therapies.
Collapse
Affiliation(s)
- Sithara Ramdas
- MDUK Neuromuscular centre, Children's Hospital, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - David Beeson
- Neurosciences Group, Weatherall Institute of Molecular Medicine, The John Radcliffe, Oxford OX3 9DS, UK.
| |
Collapse
|
33
|
Kingwell K. Docking downstream to treat myasthenia. Nat Rev Drug Discov 2021; 20:587. [PMID: 34239091 DOI: 10.1038/d41573-021-00120-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|