1
|
Ishida M, Ichikawa R, Ohbuchi K, Oizumi H, Miyamoto Y, Yamauchi J. A tardive dyskinesia drug target VMAT-2 participates in neuronal process elongation. Sci Rep 2025; 15:12049. [PMID: 40200061 PMCID: PMC11978964 DOI: 10.1038/s41598-025-97308-5] [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: 10/31/2024] [Accepted: 04/03/2025] [Indexed: 04/10/2025] Open
Abstract
Tardive dyskinesia involves involuntary movements of body parts and is often observed in individuals taking antipsychotics for extended periods. Initial treatment strategies include reducing medication dosage, switching medications, or using drugs to suppress symptoms. One of the therapeutic targets for tardive dyskinesia is vesicular monoamine transporter-2 (VMAT-2, also known as solute carrier family 18 member A2 [SLC18A2]), which functions as an energy-dependent transporter of monoamines. The therapeutic drugs are used during adulthood, when neurons are maturing. For the first time, we report that treatment with a chemical VMAT-2 inhibitor reduces neuronal process elongation, a phenomenon commonly observed during development. Treatment with the inhibitors reserpine or tetrabenazine decreased process elongation in primary cortical neurons, and similar results were obtained in N1E-115 neuronal model cells undergoing process elongation. Knockdown of VMAT-2 using clustered regularly interspaced short palindromic repeat (CRISPR)/Cas13-fitted guide RNA also reduced process elongation. However, treatment with reserpine or tetrabenazine did not affect the morphology of mature processes. Notably, treatment with hesperetin, a citrus flavonoid with neuroprotective effects, was able to restore the reduced process elongation induced by these inhibitors or VMAT-2 knockdown. The underlying molecular mechanism appeared to involve neuronal differentiation-related Akt kinase signaling. These results suggest that VMAT-2, as a drug target for tardive dyskinesia, plays a key role in process elongation and that some inhibitory effects of VMAT-2-targeted drugs on its elongation may be mitigated by co-administering a neuroprotective molecule.
Collapse
Affiliation(s)
- Miki Ishida
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Ryuya Ichikawa
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Katsuya Ohbuchi
- Tsumura Research Laboratories, Tsumura & Co., Ibaraki, 200-1192, Japan
| | - Hiroaki Oizumi
- Tsumura Research Laboratories, Tsumura & Co., Ibaraki, 200-1192, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
- Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
- Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan.
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan.
| |
Collapse
|
2
|
Deng H, Zhang Q, Yi J, Yuan L. Unraveling ptosis: A comprehensive review of clinical manifestations, genetics, and treatment. Prog Retin Eye Res 2025; 105:101327. [PMID: 39725023 DOI: 10.1016/j.preteyeres.2024.101327] [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: 09/05/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Ptosis is defined as an abnormally low-lying upper eyelid margin on the primary gaze, generally resulting from a congenital or acquired abnormality of the nerves or muscles that control the eyelid. Ptosis can occur alone or concurrently as an ocular or systemic syndrome, and the prevalence of ptosis varies among different countries and populations. Isolated ptosis typically causes aesthetic problems in patients and can lead to functional ophthalmic problems in severe cases. In individuals with syndromic ptosis, ptosis can be a warning of serious medical problems. There are different approaches to classification, depending on the onset time or the etiology of ptosis, and the clinical characteristics of congenital and acquired ptosis also differ. Pedigree and genetic analysis have demonstrated that hereditary ptosis is clinically heterogeneous, with incomplete concordance and variable expressivity. A number of genetic loci and genes responsible for hereditary isolated and syndromic ptosis have been reported. Optimal surgical timing and proper method are truly critical for avoiding the risk of potentially severe outcomes from ptosis and minimizing surgical complications, which are challenging as the pathogenesis is still indistinct and the anatomy is complex. This review provides a comprehensive review of ptosis, by summarizing the clinical manifestations, classification, diagnosis, genetics, treatment, and prognosis, as well as the bound anatomy of upper eyelid.
Collapse
Affiliation(s)
- Hao Deng
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Research Center of Medical Experimental Technology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Disease Genome Research Center, Central South University, Changsha, 410013, China; Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Qianling Zhang
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Junhui Yi
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Lamei Yuan
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Research Center of Medical Experimental Technology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China; Disease Genome Research Center, Central South University, Changsha, 410013, China.
| |
Collapse
|
3
|
Ma Q, Ma K, Dong Y, Meng Y, Zhao J, Li R, Bai Q, Wu D, Jiang D, Sun J, Zhao Y. Binding mechanism and antagonism of the vesicular acetylcholine transporter VAChT. Nat Struct Mol Biol 2025:10.1038/s41594-024-01462-9. [PMID: 39806024 DOI: 10.1038/s41594-024-01462-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
The vesicular acetylcholine transporter (VAChT) has a pivotal role in packaging and transporting acetylcholine for exocytotic release, serving as a vital component of cholinergic neurotransmission. Dysregulation of its function can result in neurological disorders. It also serves as a target for developing radiotracers to quantify cholinergic neuron deficits in neurodegenerative conditions. Here we unveil the cryo-electron microscopy structures of human VAChT in its apo state, the substrate acetylcholine-bound state and the inhibitor vesamicol-bound state. These structures assume a lumen-facing conformation, offering a clear depiction of architecture of VAChT. The acetylcholine-bound structure provides a detailed understanding of how VAChT recognizes its substrate, shedding light on the coupling mechanism of protonation and substrate binding. Meanwhile, the vesamicol-bound structure reveals the binding mode of vesamicol to VAChT, laying the structural foundation for the design of the next generation of radioligands targeting VAChT.
Collapse
Affiliation(s)
- Qiao Ma
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kunpeng Ma
- Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yanli Dong
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yufei Meng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun Zhao
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, China
| | - Renjie Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qinru Bai
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Di Wu
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Daohua Jiang
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Jianyuan Sun
- University of Chinese Academy of Sciences, Beijing, China
- Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yan Zhao
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Nagy MA, Price S, Wang K, Gill S, Ren E, Jayne L, Pajak V, Deighan S, Liu B, Lu X, Diallo A, Lo SC, Kleiman R, Henderson C, Suh J, Griffith EC, Greenberg ME, Hrvatin S. Cis-regulatory elements driving motor neuron-selective viral payload expression within the mammalian spinal cord. Proc Natl Acad Sci U S A 2024; 121:e2418024121. [PMID: 39602276 PMCID: PMC11626145 DOI: 10.1073/pnas.2418024121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Spinal motor neuron (MN) dysfunction is the cause of a number of clinically significant movement disorders. Despite the recent approval of gene therapeutics targeting these MN-related disorders, there are no viral delivery mechanisms that achieve MN-restricted transgene expression. In this study, chromatin accessibility profiling of genetically defined mouse MNs was used to identify candidate cis-regulatory elements (CREs) capable of driving MN-selective gene expression. Subsequent testing of these candidates identified two CREs that confer MN-selective gene expression in the spinal cord as well as reduced off-target expression in dorsal root ganglia. Within one of these candidate elements, we identified a compact core transcription factor (TF)-binding region that drives MN-selective gene expression. Finally, we demonstrated that selective spinal cord expression driven by this mouse CRE is preserved in non-human primates. These findings suggest that cell-type-selective viral reagents in which cell-type-selective CREs drive restricted gene expression will be valuable research tools in mice and other mammalian species, with potentially significant therapeutic value in humans.
Collapse
Affiliation(s)
- M. Aurel Nagy
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Harvard Medical School, Boston, MA02115
| | - Spencer Price
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Kristina Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
- Department of Immunology, Harvard Medical School, Boston, MA02115
| | - Stanley Gill
- Harvard Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Erika Ren
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Lorna Jayne
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Victoria Pajak
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Sarah Deighan
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | | | | | | | | | | | | | - Eric C. Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | - Sinisa Hrvatin
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
5
|
Rossi L, Mota BI, Valadão PAC, Magalhães-Gomes MPS, Oliveira BS, Guatimosim S, Navegantes LCC, Miranda AS, Prado MAM, Prado VF, Guatimosim C. Influence of β 2-adrenergic selective agonist formoterol on the motor unit of a mouse model of a congenital myasthenic syndrome with complete VAChT deletion. Neuropharmacology 2024; 260:110116. [PMID: 39151654 DOI: 10.1016/j.neuropharm.2024.110116] [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: 05/05/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Congenital Myasthenic Syndromes (CMS) are a set of genetic diseases that affect the neuromuscular transmission causing muscular weakness. The standard pharmacological treatment aims at ameliorating the myasthenic symptom by acetylcholinesterase inhibitors. Most patients respond well in the short and medium term, however, over time the beneficial effects rapidly fade, and the efficacy of the treatment diminishes. Increasing evidence shows that β2-adrenergic agonists can be a suitable choice for the treatment of neuromuscular disorders, including CMS, as they promote beneficial effects in the neuromuscular system. The exact mechanism on which they rely is not completely understood, although patients and animal models respond well to the treatment, especially over extended periods. Here, we report the use of the long-lasting specific β2-adrenergic agonist formoterol in a myasthenic mouse model (mnVAChT-KD), featuring deletion of VAChT (Vesicular Acetylcholine Transporter) specifically in the α-motoneurons. Our findings demonstrate that formoterol treatment (300 μg/kg/day; sc) for 30 days increased the neuromuscular junction area, induced skeletal muscle hypertrophy and altered fibre type composition in myasthenic mice. Interestingly, β2-adrenergic agonists have shown efficacy even in the absence of ACh (acetylcholine). Our data provide important evidence supporting the potential of β2-adrenergic agonists in treating neuromuscular disorders of pre-synaptic origin and characterized by disruptions in nerve-muscle communication, through a direct and beneficial action within the motor unit.
Collapse
Affiliation(s)
- Leonardo Rossi
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bárbara I Mota
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Priscila A C Valadão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Matheus P S Magalhães-Gomes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Departamento de Ciências Básicas, Faculdade Ciências Médicas de Minas Gerais, FCMMG, Belo Horizonte, Brazil
| | - Bruna S Oliveira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Silvia Guatimosim
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz C C Navegantes
- Departamento de Fisiologia, Escola de Medicina, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Aline S Miranda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marco A M Prado
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada; Brain and Mind Institute, University of Western Ontario, London, Canada
| | - Vânia F Prado
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada; Brain and Mind Institute, University of Western Ontario, London, Canada
| | - Cristina Guatimosim
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
6
|
Zhang Y, Dai F, Chen N, Zhou D, Lee CH, Song C, Zhang Y, Zhang Z. Structural insights into VAChT neurotransmitter recognition and inhibition. Cell Res 2024; 34:665-668. [PMID: 38862671 PMCID: PMC11369146 DOI: 10.1038/s41422-024-00986-5] [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: 03/29/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024] Open
Affiliation(s)
- Yang Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Fei Dai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Nanhao Chen
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Dong Zhou
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Chen Song
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | - Yixiao Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| | - Zhe Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China.
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
7
|
Muntadas JA, Hyland MR, Martínez MDRO, Young JN, Chong JX, Bamshad MJ, Maselli RA. Congenital myasthenic syndrome secondary to pathogenic variants in the SLC5A7 gene: report of two cases. BMC Med Genomics 2024; 17:207. [PMID: 39135055 PMCID: PMC11318227 DOI: 10.1186/s12920-024-01977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Congenital Myasthenic Syndromes (CMS) are rare genetic diseases, which share as a common denominator muscle fatigability due to failure of neuromuscular transmission. A distinctive clinical feature of presynaptic CMS variants caused by defects of the synthesis of acetylcholine is the association with life-threatening episodes of apnea. One of these variants is caused by mutations in the SLC5A7 gene, which encodes the sodium-dependent HC-3 high-affinity choline transporter 1 (CHT1). To our knowledge there are no published cases of this CMS type in Latin America. CASE PRESENTATION We present two cases of CHT1-CMS. Both patients were males presenting with repeated episodes of apnea, hypotonia, weakness, ptosis, mild ophthalmoparesis, and bulbar deficit. The first case also presented one isolated seizure, while the second case showed global developmental delay. Both cases, exhibited incomplete improvement with treatment with pyridostigmine. CONCLUSIONS This report emphasizes the broad incidence of CMS with episodic apnea caused by mutations in the SLC5A7 gene and the frequent association of this condition with serious manifestations of central nervous system involvement.
Collapse
Affiliation(s)
- Javier A Muntadas
- Pediatric Neurology, Hospital Italiano, Gascon 450. Capital Federal, Buenos Aires, 4959-0200, Argentina
| | - Martin R Hyland
- Pediatric Neurology, Hospital Italiano, Gascon 450. Capital Federal, Buenos Aires, 4959-0200, Argentina
| | | | - Jaime N Young
- Department of Neurology, University of California Davis, 1515 Newton Court, Davis, CA, 95618, USA
| | - Jessica X Chong
- Brotman Baty Institute for Precision Medicine, 1959 NE Pacific St, Seattle, WA, 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, 4245 Roosevelt Way NE, Seattle, WA, 98105, USA
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, William H. Foege Hall, 3720 15th Ave NE, Seattle, WA, 98195, USA
- Brotman Baty Institute for Precision Medicine, 1959 NE Pacific St, Seattle, WA, 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, 4245 Roosevelt Way NE, Seattle, WA, 98105, USA
| | - Ricardo A Maselli
- Department of Neurology, University of California Davis, 1515 Newton Court, Davis, CA, 95618, USA.
| |
Collapse
|
8
|
Ji B, Chen J, Gong H, Li X. Streamlined Full-Length Total RNA Sequencing of Paraformaldehyde-Fixed Brain Tissues. Int J Mol Sci 2024; 25:6504. [PMID: 38928210 PMCID: PMC11204141 DOI: 10.3390/ijms25126504] [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: 04/25/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Paraformaldehyde (PFA) fixation is the preferred method for preserving tissue architecture for anatomical and pathological observations. Meanwhile, PFA reacts with the amine groups of biomolecules to form chemical cross-linking, which preserves RNA within the tissue. This has great prospects for RNA sequencing to characterize the molecular underpinnings after anatomical and pathological observations. However, RNA is inaccessible due to cross-linked adducts forming between RNA and other biomolecules in prolonged PFA-fixed tissue. It is also difficult to perform reverse transcription and PCR, resulting in low sequencing sensitivity and reduced reproducibility. Here, we developed a method to perform RNA sequencing in PFA-fixed tissue, which is easy to use, cost-effective, and allows efficient sample multiplexing. We employ cross-link reversal to recover RNA and library construction using random primers without artificial fragmentation. The yield and quality of recovered RNA significantly increased through our method, and sequencing quality metrics and detected genes did not show any major differences compared with matched fresh samples. Moreover, we applied our method for gene expression analysis in different regions of the mouse brain and identified unique gene expression profiles with varied functional implications. We also find significant dysregulation of genes involved in Alzheimer's disease (AD) pathogenesis within the medial septum (MS)/vertical diagonal band of Broca (VDB) of the 5×FAD mouse brain. Our method can thus increase the performance of high-throughput RNA sequencing with PFA-fixed samples and allows longitudinal studies of small tissue regions isolated by their in situ context.
Collapse
Affiliation(s)
- Bingqing Ji
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (B.J.); (J.C.); (H.G.)
- MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiale Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (B.J.); (J.C.); (H.G.)
- MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China; (B.J.); (J.C.); (H.G.)
- MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215125, China
| | - Xiangning Li
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215125, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
9
|
Marchant RG, Bryen SJ, Bahlo M, Cairns A, Chao KR, Corbett A, Davis MR, Ganesh VS, Ghaoui R, Jones KJ, Kornberg AJ, Lek M, Liang C, MacArthur DG, Oates EC, O'Donnell-Luria A, O'Grady GL, Osei-Owusu IA, Rafehi H, Reddel SW, Roxburgh RH, Ryan MM, Sandaradura SA, Scott LW, Valkanas E, Weisburd B, Young H, Evesson FJ, Waddell LB, Cooper ST. Genome and RNA sequencing boost neuromuscular diagnoses to 62% from 34% with exome sequencing alone. Ann Clin Transl Neurol 2024; 11:1250-1266. [PMID: 38544359 DOI: 10.1002/acn3.52041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/24/2024] [Indexed: 05/15/2024] Open
Abstract
OBJECTIVE Most families with heritable neuromuscular disorders do not receive a molecular diagnosis. Here we evaluate diagnostic utility of exome, genome, RNA sequencing, and protein studies and provide evidence-based recommendations for their integration into practice. METHODS In total, 247 families with suspected monogenic neuromuscular disorders who remained without a genetic diagnosis after standard diagnostic investigations underwent research-led massively parallel sequencing: neuromuscular disorder gene panel, exome, genome, and/or RNA sequencing to identify causal variants. Protein and RNA studies were also deployed when required. RESULTS Integration of exome sequencing and auxiliary genome, RNA and/or protein studies identified causal or likely causal variants in 62% (152 out of 247) of families. Exome sequencing alone informed 55% (83 out of 152) of diagnoses, with remaining diagnoses (45%; 69 out of 152) requiring genome sequencing, RNA and/or protein studies to identify variants and/or support pathogenicity. Arrestingly, novel disease genes accounted for <4% (6 out of 152) of diagnoses while 36.2% of solved families (55 out of 152) harbored at least one splice-altering or structural variant in a known neuromuscular disorder gene. We posit that contemporary neuromuscular disorder gene-panel sequencing could likely provide 66% (100 out of 152) of our diagnoses today. INTERPRETATION Our results emphasize thorough clinical phenotyping to enable deep scrutiny of all rare genetic variation in phenotypically consistent genes. Post-exome auxiliary investigations extended our diagnostic yield by 81% overall (34-62%). We present a diagnostic algorithm that details deployment of genomic and auxiliary investigations to obtain these diagnoses today most effectively. We hope this provides a practical guide for clinicians as they gain greater access to clinical genome and transcriptome sequencing.
Collapse
Affiliation(s)
- Rhett G Marchant
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Samantha J Bryen
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Melanie Bahlo
- Functional Neuromics, Children's Medical Research Institute, Westmead, New South Wales, Australia
- Population Health and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Anita Cairns
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Neurosciences Department, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Katherine R Chao
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Alastair Corbett
- Neurology Department, Repatriation General Hospital Concord, Concord, New South Wales, Australia
| | - Mark R Davis
- Department of Diagnostic Genomics, PathWest Laboratory Medicine, Perth, WA, Australia
| | - Vijay S Ganesh
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Neuromuscular Division, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Roula Ghaoui
- Department of Neurology, Central Adelaide Local Health Network/Royal Adelaide Hospital, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Genetics & Molecular Pathology, SA Pathology, Adelaide, South Australia, Australia
| | - Kristi J Jones
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Clinical Genetics, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Andrew J Kornberg
- Department of Neurology, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Monkol Lek
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Liang
- Department of Neurology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Neurogenetics, Northern Clinical School, Kolling Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Daniel G MacArthur
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Centre for Population Genomics, Garvan Institute of Medical Research/University of New South Wales, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Emily C Oates
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Randwick, New South Wales, Australia
| | - Anne O'Donnell-Luria
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gina L O'Grady
- Starship Children's Health, Auckland District Health Board, Auckland, New Zealand
| | - Ikeoluwa A Osei-Owusu
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Haloom Rafehi
- Functional Neuromics, Children's Medical Research Institute, Westmead, New South Wales, Australia
- Population Health and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Stephen W Reddel
- Neurology Department, Repatriation General Hospital Concord, Concord, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard H Roxburgh
- Department of Neurology, Auckland District Health Board, Auckland, New Zealand
- Centre of Brain Research Neurogenetics Research Clinic, University of Auckland, Auckland, New Zealand
| | - Monique M Ryan
- Department of Neurology, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Sarah A Sandaradura
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Clinical Genetics, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Liam W Scott
- Functional Neuromics, Children's Medical Research Institute, Westmead, New South Wales, Australia
- Population Health and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Elise Valkanas
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Ben Weisburd
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Helen Young
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Department of Neurology, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Paediatrics, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Frances J Evesson
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Leigh B Waddell
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Sandra T Cooper
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
10
|
Liao K, Xiang Y, Huang F, Huang M, Xu W, Lin Y, Liao P, Wang Z, Yang L, Tian X, Chen D, Wang Z, Liu S, Zhuang Z. Spatial and single-nucleus transcriptomics decoding the molecular landscape and cellular organization of avian optic tectum. iScience 2024; 27:109009. [PMID: 38333704 PMCID: PMC10850779 DOI: 10.1016/j.isci.2024.109009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
The avian optic tectum (OT) has been studied for its diverse functions, yet a comprehensive molecular landscape at the cellular level has been lacking. In this study, we applied spatial transcriptome sequencing and single-nucleus RNA sequencing (snRNA-seq) to explore the cellular organization and molecular characteristics of the avian OT from two species: Columba livia and Taeniopygia guttata. We identified precise layer structures and provided comprehensive layer-specific signatures of avian OT. Furthermore, we elucidated diverse functions in different layers, with the stratum griseum periventriculare (SGP) potentially playing a key role in advanced functions of OT, like fear response and associative learning. We characterized detailed neuronal subtypes and identified a population of FOXG1+ excitatory neurons, resembling those found in the mouse neocortex, potentially involved in neocortex-related functions and expansion of avian OT. These findings could contribute to our understanding of the architecture of OT, shedding light on visual perception and multifunctional association.
Collapse
Affiliation(s)
- Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
- BGI Research, Hangzhou 310030, China
| | - Ya Xiang
- BGI Research, Hangzhou 310030, China
- College of Life Sciences, Northwest University, Xi’an 710069, China
| | - Fubaoqian Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
- BGI Research, Hangzhou 310030, China
| | - Maolin Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenbo Xu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Youning Lin
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Pingfang Liao
- BGI Research, Hangzhou 310030, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zishi Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lin Yang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xinmao Tian
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Duoyuan Chen
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Zhenlong Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Zhenkun Zhuang
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| |
Collapse
|
11
|
Lin CV, Thomas CAD, Huynh TL, Wei DT, Young JN, Aivazian AS, McInnes A, Xu J, Cook SE, Vazquez J, Maselli RA. Adeno-Associated Virus Type 9-Mediated Gene Therapy of Choline Acetyltransferase-Deficient Mice. Hum Gene Ther 2024; 35:123-131. [PMID: 38299967 DOI: 10.1089/hum.2023.173] [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] [Indexed: 02/02/2024] Open
Abstract
The enzyme choline acetyltransferase (ChAT) synthesizes acetylcholine from acetyl-CoA and choline at the neuromuscular junction and at the nerve terminals of cholinergic neurons. Mutations in the ChAT gene (CHAT) result in a presynaptic congenital myasthenic syndrome (CMS) that often associates with life-threatening episodes of apnea. Knockout mice for Chat (Chat-/-) die at birth. To circumvent the lethality of this model, we crossed mutant mice possessing loxP sites flanking Chat exons 4 and 5 with mice that expressed Cre-ERT2. Injection of tamoxifen (Tx) at postnatal (P) day 11 in these mice induced downregulation of Chat, autonomic failure, weakness, and death. However, a proportion of Chatflox/flox-Cre-ERT2 mice receiving at birth an intracerebroventricular injection of 2 × 1013 vg/kg adeno-associated virus type 9 (AAV9) carrying human CHAT (AAV9-CHAT) survived a subsequent Tx injection and lived to adulthood without showing signs of weakness. Likewise, injection of AA9-CHAT by intracisternal injection at P28 after the onset of weakness also resulted in survival to adulthood. The expression of Chat in spinal motor neurons of Chatflox/flox-Cre-ERT2 mice injected with Tx was markedly reduced, but AAV-injected mice showed a robust recovery of ChAT expression, which was mainly translated by the human CHAT RNA. The biodistribution of the viral genome was widespread but maximal in the spinal cord and brain of AAV-injected mice. No significant histopathological changes were observed in the brain, liver, and heart of AAV-injected mice after 1 year follow-up. Thus, AAV9-mediated gene therapy may provide an effective and safe treatment for patients severely affected with CHAT-CMS.
Collapse
Affiliation(s)
- Cameron V Lin
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Clementine A D Thomas
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Thanh L Huynh
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - David T Wei
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Jaime N Young
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Anahid S Aivazian
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Abigail McInnes
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Jixiang Xu
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Sarah E Cook
- Anatomic Pathology, University of California Davis, School of Veterinary Medicine, Davis, California, USA
| | - Jessica Vazquez
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| | - Ricardo A Maselli
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, California, USA
| |
Collapse
|
12
|
Erbe LS, Hoffjan S, Janßen S, Kneifel M, Krause K, Gerding WM, Döring K, Güttsches AK, Roos A, Buena Atienza E, Gross C, Lücke T, Nguyen HHP, Vorgerd M, Köhler C. Exome Sequencing and Optical Genome Mapping in Molecularly Unsolved Cases of Duchenne Muscular Dystrophy: Identification of a Causative X-Chromosomal Inversion Disrupting the DMD Gene. Int J Mol Sci 2023; 24:14716. [PMID: 37834164 PMCID: PMC10572545 DOI: 10.3390/ijms241914716] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe progressive muscle disease that mainly affects boys due to X-linked recessive inheritance. In most affected individuals, MLPA or sequencing-based techniques detect deletions, duplications, or point mutations in the dystrophin-encoding DMD gene. However, in a small subset of patients clinically diagnosed with DMD, the molecular cause is not identified with these routine methods. Evaluation of the 60 DMD patients in our center revealed three cases without a known genetic cause. DNA samples of these patients were analyzed using whole-exome sequencing (WES) and, if unconclusive, optical genome mapping (OGM). WES led to a diagnosis in two cases: one patient was found to carry a splice mutation in the DMD gene that had not been identified during previous Sanger sequencing. In the second patient, we detected two variants in the fukutin gene (FKTN) that were presumed to be disease-causing. In the third patient, WES was unremarkable, but OGM identified an inversion disrupting the DMD gene (~1.28 Mb) that was subsequently confirmed with long-read sequencing. These results highlight the importance of reanalyzing unsolved cases using WES and demonstrate that OGM is a useful method for identifying large structural variants in cases with unremarkable exome sequencing.
Collapse
Affiliation(s)
- Leoni S. Erbe
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
| | - Sabine Hoffjan
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
| | - Sören Janßen
- Department of Neuropediatrics, University Children’s Hospital, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Moritz Kneifel
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Karsten Krause
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Wanda M. Gerding
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
| | - Kristina Döring
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
| | - Anne-Katrin Güttsches
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Andreas Roos
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Elena Buena Atienza
- Institute of Medical Genetics and Applied Genomics, University Tübingen, 72074 Tübingen, Germany; (E.B.A.); (C.G.)
- NGS Competence Center Tübingen, 72076 Tübingen, Germany
| | - Caspar Gross
- Institute of Medical Genetics and Applied Genomics, University Tübingen, 72074 Tübingen, Germany; (E.B.A.); (C.G.)
- NGS Competence Center Tübingen, 72076 Tübingen, Germany
| | - Thomas Lücke
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
- Department of Neuropediatrics, University Children’s Hospital, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Hoa Huu Phuc Nguyen
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Cornelia Köhler
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
- Department of Neuropediatrics, University Children’s Hospital, Ruhr-University Bochum, 44801 Bochum, Germany;
| |
Collapse
|
13
|
Anchesi I, Schepici G, Chiricosta L, Gugliandolo A, Salamone S, Caprioglio D, Pollastro F, Mazzon E. Δ 8-THC Induces Up-Regulation of Glutamatergic Pathway Genes in Differentiated SH-SY5Y: A Transcriptomic Study. Int J Mol Sci 2023; 24:ijms24119486. [PMID: 37298437 DOI: 10.3390/ijms24119486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Cannabinoids, natural or synthetic, have antidepressant, anxiolytic, anticonvulsant, and anti-psychotic properties. Cannabidiol (CBD) and delta-9-tetrahydrocannabinol (Δ9-THC) are the most studied cannabinoids, but recently, attention has turned towards minor cannabinoids. Delta-8-tetrahydrocannabinol (Δ8-THC), an isomer of Δ9-THC, is a compound for which, to date, there is no evidence of its role in the modulation of synaptic pathways. The aim of our work was to evaluate the effects of Δ8-THC on differentiated SH-SY5Y human neuroblastoma cells. Using next generation sequencing (NGS), we investigated whether Δ8-THC could modify the transcriptomic profile of genes involved in synapse functions. Our results showed that Δ8-THC upregulates the expression of genes involved in the glutamatergic pathway and inhibits gene expression at cholinergic synapses. Conversely, Δ8-THC did not modify the transcriptomic profile of genes involved in the GABAergic and dopaminergic pathways.
Collapse
Affiliation(s)
- Ivan Anchesi
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Giovanni Schepici
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Diego Caprioglio
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
14
|
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: 54] [Impact Index Per Article: 27.0] [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
|
15
|
Pugliese A, Holland SH, Rodolico C, Lochmüller H, Spendiff S. Presynaptic Congenital Myasthenic Syndromes: Understanding Clinical Phenotypes through In vivo Models. J Neuromuscul Dis 2023; 10:731-759. [PMID: 37212067 PMCID: PMC10578258 DOI: 10.3233/jnd-221646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2023] [Indexed: 05/23/2023]
Abstract
Presynaptic congenital myasthenic syndromes (CMS) are a group of genetic disorders affecting the presynaptic side of the neuromuscular junctions (NMJ). They can result from a dysfunction in acetylcholine (ACh) synthesis or recycling, in its packaging into synaptic vesicles, or its subsequent release into the synaptic cleft. Other proteins involved in presynaptic endplate development and maintenance can also be impaired.Presynaptic CMS usually presents during the prenatal or neonatal period, with a severe phenotype including congenital arthrogryposis, developmental delay, and apnoeic crisis. However, milder phenotypes with proximal muscle weakness and good response to treatment have been described. Finally, many presynaptic genes are expressed in the brain, justifying the presence of additional central nervous system symptoms.Several animal models have been developed to study CMS, providing the opportunity to identify disease mechanisms and test treatment options. In this review, we describe presynaptic CMS phenotypes with a focus on in vivo models, to better understand CMS pathophysiology and define new causative genes.
Collapse
Affiliation(s)
- Alessia Pugliese
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Stephen H. Holland
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Carmelo Rodolico
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Hanns Lochmüller
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, 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
| | - Sally Spendiff
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| |
Collapse
|
16
|
Kong W, Montano M, Corley MJ, Helmy E, Kobayashi H, Kinisu M, Suryawanshi R, Luo X, Royer LA, Roan NR, Ott M, Ndhlovu LC, Greene WC. Neuropilin-1 Mediates SARS-CoV-2 Infection of Astrocytes in Brain Organoids, Inducing Inflammation Leading to Dysfunction and Death of Neurons. mBio 2022; 13:e0230822. [PMID: 36314791 PMCID: PMC9765283 DOI: 10.1128/mbio.02308-22] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/05/2022] [Indexed: 11/07/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is frequently associated with neurological deficits, but how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces these effects remains unclear. Here, we show that astrocytes are readily infected by SARS-CoV-2, but surprisingly, neuropilin-1, not angiotensin-converting enzyme 2 (ACE2), serves as the principal receptor mediating cell entry. Infection is further positively modulated by the two-pore segment channel 2 (TPC2) protein that regulates membrane trafficking and endocytosis. Astrocyte infection produces a pathological response closely resembling reactive astrogliosis characterized by elevated type I interferon (IFN) production, increased inflammation, and the decreased expression of transporters of water, ions, choline, and neurotransmitters. These combined events initiated within astrocytes produce a hostile microenvironment that promotes the dysfunction and death of uninfected bystander neurons. IMPORTANCE SARS-CoV-2 infection primarily targets the lung but may also damage other organs, including the brain, heart, kidney, and intestine. Central nervous system (CNS) pathologies include loss of smell and taste, headache, delirium, acute psychosis, seizures, and stroke. Pathological loss of gray matter occurs in SARS-CoV-2 infection, but it is unclear whether this is due to direct viral infection, indirect effects associated with systemic inflammation, or both. Here, we used induced pluripotent stem cell (iPSC)-derived brain organoids and primary human astrocytes from the cerebral cortex to study direct SARS-CoV-2 infection. Our findings support a model where SARS-CoV-2 infection of astrocytes produces a panoply of changes in the expression of genes regulating innate immune signaling and inflammatory responses. The deregulation of these genes in astrocytes produces a microenvironment within the CNS that ultimately disrupts normal neuron function, promoting neuronal cell death and CNS deficits.
Collapse
Affiliation(s)
- Weili Kong
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
| | - Mauricio Montano
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
| | - Michael J. Corley
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Ekram Helmy
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
| | | | - Martin Kinisu
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
| | | | - Xiaoyu Luo
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
| | | | - Nadia R. Roan
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
- Department of Urology, University of California, San Francisco, San Francisco, California, USA
| | - Melanie Ott
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, USA
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Warner C. Greene
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, California, USA
- Gladstone Institute of Virology, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
17
|
Francesca P, Mauro P, Clerbaux LA, Leoni G, Ponti J, Bogni A, Brogna C, Cristoni S, Sanges R, Mendoza-de Gyves E, Fabbri M, Querci M, Soares H, Munoz Pineiro A, Whelan M, Van de Eede G. Effects of spike protein and toxin-like peptides found in COVID-19 patients on human 3D neuronal/glial model undergoing differentiation: possible implications for SARS-CoV-2 impact on brain development. Reprod Toxicol 2022; 111:34-48. [PMID: 35525527 PMCID: PMC9068247 DOI: 10.1016/j.reprotox.2022.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/28/2022] [Accepted: 04/30/2022] [Indexed: 12/13/2022]
Abstract
The possible neurodevelopmental consequences of SARS-CoV-2 infection are presently unknown. In utero exposure to SARS-CoV-2 has been hypothesized to affect the developing brain, possibly disrupting neurodevelopment of children. Spike protein interactors, such as ACE2, have been found expressed in the fetal brain, and could play a role in potential SARS-CoV-2 fetal brain pathogenesis. Apart from the possible direct involvement of SARS-CoV-2 or its specific viral components in the occurrence of neurological and neurodevelopmental manifestations, we recently reported the presence of toxin-like peptides in plasma, urine and fecal samples specifically from COVID-19 patients. In this study, we investigated the possible neurotoxic effects elicited upon 72-hour exposure to human relevant levels of recombinant spike protein, toxin-like peptides found in COVID-19 patients, as well as a combination of both in 3D human iPSC-derived neural stem cells differentiated for either 2 weeks (short-term) or 8 weeks (long-term, 2 weeks in suspension + 6 weeks on MEA) towards neurons/glia. Whole transcriptome and qPCR analysis revealed that spike protein and toxin-like peptides at non-cytotoxic concentrations differentially perturb the expression of SPHK1, ELN, GASK1B, HEY1, UTS2, ACE2 and some neuronal-, glia- and NSC-related genes critical during brain development. Additionally, exposure to spike protein caused a decrease of spontaneous electrical activity after two days in long-term differentiated cultures. The perturbations of these neurodevelopmental endpoints are discussed in the context of recent knowledge about the key events described in Adverse Outcome Pathways relevant to COVID-19, gathered in the context of the CIAO project (https://www.ciao-covid.net/).
Collapse
Affiliation(s)
| | - Petrillo Mauro
- Seidor Italy srl. Past affiliation (until 15/06/2021) European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Gabriele Leoni
- European Commission, Joint Research Centre (JRC), Ispra, Italy; International School for Advanced Studies (SISSA), Trieste, Italy
| | - Jessica Ponti
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Alessia Bogni
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | - Remo Sanges
- International School for Advanced Studies (SISSA), Trieste, Italy
| | | | - Marco Fabbri
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Helena Soares
- Human Immunobiology and Pathogenesis Group, CEDOC, NOVA Medical School
- Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | | | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Guy Van de Eede
- European Commission, Joint Research Centre (JRC), Geel, Belgium
| |
Collapse
|
18
|
Della Marina A, Arlt A, Schara-Schmidt U, Depienne C, Gangfuß A, Kölbel H, Sickmann A, Freier E, Kohlschmidt N, Hentschel A, Weis J, Czech A, Grüneboom A, Roos A. Phenotypical and Myopathological Consequences of Compound Heterozygous Missense and Nonsense Variants in SLC18A3. Cells 2021; 10:cells10123481. [PMID: 34943989 PMCID: PMC8700530 DOI: 10.3390/cells10123481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Presynaptic forms of congenital myasthenic syndromes (CMS) due to pathogenic variants in SLC18A3 impairing the synthesis and recycling of acetylcholine (ACh) have recently been described. SLC18A3 encodes the vesicular ACh transporter (VAChT), modulating the active transport of ACh at the neuromuscular junction, and homozygous loss of VAChT leads to lethality. Methods: Exome sequencing (ES) was carried out to identify the molecular genetic cause of the disease in a 5-year-old male patient and histological, immunofluorescence as well as electron- and CARS-microscopic studies were performed to delineate the muscle pathology, which has so far only been studied in VAChT-deficient animal models. Results: ES unraveled compound heterozygous missense and nonsense variants (c.315G>A, p.Trp105* and c.1192G>C, p.Asp398His) in SLC18A3. Comparison with already-published cases suggests a more severe phenotype including impaired motor and cognitive development, possibly related to a more severe effect of the nonsense variant. Therapy with pyridostigmine was only partially effective while 3,4 diaminopyridine showed no effect. Microscopic investigation of the muscle biopsy revealed reduced fibre size and a significant accumulation of lipid droplets. Conclusions: We suggest that nonsense variants have a more detrimental impact on the clinical manifestation of SLC18A3-associated CMS. The impact of pathogenic SLC18A3 variants on muscle fibre integrity beyond the effect of denervation is suggested by the build-up of lipid aggregates. This in turn implicates the importance of proper VAChT-mediated synthesis and recycling of ACh for lipid homeostasis in muscle cells. This hypothesis is further supported by the pathological observations obtained in previously published VAChT-animal models.
Collapse
Affiliation(s)
- Adela Della Marina
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (U.S.-S.); (A.G.); (H.K.); (A.R.)
- Correspondence:
| | - Annabelle Arlt
- Institute of Clinical Genetics and Tumor Genetics Bonn, 53111 Bonn, Germany; (A.A.); (N.K.)
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (U.S.-S.); (A.G.); (H.K.); (A.R.)
| | - Christel Depienne
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany;
| | - Andrea Gangfuß
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (U.S.-S.); (A.G.); (H.K.); (A.R.)
| | - Heike Kölbel
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (U.S.-S.); (A.G.); (H.K.); (A.R.)
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., 44139 Dortmund, Germany; (A.S.); (E.F.); (A.H.); (A.C.); (A.G.)
| | - Erik Freier
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., 44139 Dortmund, Germany; (A.S.); (E.F.); (A.H.); (A.C.); (A.G.)
| | - Nicolai Kohlschmidt
- Institute of Clinical Genetics and Tumor Genetics Bonn, 53111 Bonn, Germany; (A.A.); (N.K.)
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., 44139 Dortmund, Germany; (A.S.); (E.F.); (A.H.); (A.C.); (A.G.)
| | - Joachim Weis
- Institute of Neuropathology, University Hospital Aachen, RWTH-Aachen University, 52074 Aachen, Germany;
| | - Artur Czech
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., 44139 Dortmund, Germany; (A.S.); (E.F.); (A.H.); (A.C.); (A.G.)
| | - Anika Grüneboom
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., 44139 Dortmund, Germany; (A.S.); (E.F.); (A.H.); (A.C.); (A.G.)
| | - Andreas Roos
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (U.S.-S.); (A.G.); (H.K.); (A.R.)
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
19
|
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: 26] [Impact Index Per Article: 6.5] [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
|
20
|
Joviano-Santos JV, Kljakic O, Magalhães-Gomes MPS, Valadão PAC, de Oliveira LR, Prado MAM, Prado VF, Guatimosim C. Motoneuron-specific loss of VAChT mimics neuromuscular defects seen in congenital myasthenic syndrome. FEBS J 2021; 288:5331-5349. [PMID: 33730374 DOI: 10.1111/febs.15825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 03/03/2021] [Accepted: 03/16/2021] [Indexed: 11/28/2022]
Abstract
Motoneurons (MNs) control muscle activity by releasing the neurotransmitter acetylcholine (ACh) at the level of neuromuscular junctions. ACh is packaged into synaptic vesicles by the vesicular ACh transporter (VAChT), and disruptions in its release can impair muscle contraction, as seen in congenital myasthenic syndromes (CMS). Recently, VAChT gene mutations were identified in humans displaying varying degrees of myasthenia. Moreover, mice with a global deficiency in VAChT expression display several characteristics of CMS. Despite these findings, little is known about how a long-term decrease in VAChT expression in vivo affects MNs structure and function. Using Cre-loxP technology, we generated a mouse model where VAChT is deleted in select groups of MNs (mnVAChT-KD). Molecular analysis revealed that the VAChT deletion was specific to MNs and affected approximately 50% of its population in the brainstem and spinal cord, with alpha-MNs primarily targeted (70% in spinal cord). Within each animal, the cell body area of VAChT-deleted MNs was significantly smaller compared to MNs with VAChT preserved. Likewise, muscles innervated by VAChT-deleted MNs showed atrophy while muscles innervated by VAChT-containing neurons appeared normal. In addition, mnVAChT KD mice had decreased muscle strength, were hypoactive, leaner and exhibited kyphosis. This neuromuscular dysfunction was evident at 2 months of age and became progressively worse by 6 months. Treatment of mutants with a cholinesterase inhibitor was able to improve some of the motor deficits. As these observations mimic what is seen in CMS, this new line could be valuable for assessing the efficacy of potential CMS drugs.
Collapse
Affiliation(s)
- Julliane V Joviano-Santos
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ornela Kljakic
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada
| | - Matheus P S Magalhães-Gomes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Medicina, Faculdade Ciências Médicas de Minas Gerais, FCMMG, Belo Horizonte, Brasil
| | - Priscila Aparecida C Valadão
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo R de Oliveira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marco A M Prado
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Brain and Mind Institute, University of Western Ontario, London, Canada
| | - Vania F Prado
- Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Canada.,Brain and Mind Institute, University of Western Ontario, London, Canada
| | - Cristina Guatimosim
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
21
|
Magalhães-Gomes MPS, Camargos W, Valadão PAC, Garcias RS, Rodrigues HA, Andrade JN, Teixeira VP, Naves LA, Cavalcante WLG, Gallaci M, Guatimosim S, Prado VF, Prado MAM, Guatimosim C. Increased Cholinergic Tone Causes Pre-synaptic Neuromuscular Degeneration and is Associated with Impaired Diaphragm Function. Neuroscience 2021; 460:31-42. [PMID: 33548369 DOI: 10.1016/j.neuroscience.2020.12.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 11/25/2022]
Abstract
In vertebrates, muscle activity is dependent on acetylcholine (ACh) released from neuromuscular junctions (NMJs), and changes in cholinergic neurotransmission are linked to a variety of neuromuscular diseases, including congenital myasthenic syndromes (CMS). The storage and release of ACh depends on the activity of the Vesicular Acetylcholine Transporter (VAChT), a rate-limiting step for cholinergic neurotransmission whose loss of function mutations was shown to cause human congenital myasthenia. However, we know much less about increased VAChT activity, due to copy number variations, for example. Therefore, here we investigated the impact of increased VAChT expression and consequently ACh levels at the synaptic cleft of the diaphragm NMJs. We analyzed structure and function of nerve and muscles from a mouse model of cholinergic hyperfunction (ChAT-ChR2-EYFP) with increased expression of VAChT. Our results showed a significant increase of ACh released under evoked stimuli. However, we observed deleterious changes in synaptic vesicles cycle (impaired endocytosis and decrease in vesicles number), together with structural alterations of NMJs. Interestingly, ultrastructure analyses showed that synaptic vesicles from ChAT-ChR2-EYFP mice NMJs were larger, which might be related to increased ACh load. We also observed that these larger synaptic vesicles were less rounded in comparison with control. Finally, we showed that ChAT-ChR2-EYFP mice NMJs have compromised safety factor, possible due to the structural alterations we described. These findings reveal that physiological cholinergic activity is important to maintain the structure and function of the neuromuscular system and help to understand some of the neuromuscular adverse effects experienced by chronically increased NMJ neurotransmission, such as individuals treated with cholinesterase inhibitors.
Collapse
Affiliation(s)
- Matheus P S Magalhães-Gomes
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Departamento de Medicina, Faculdade Ciências Médicas de Minas Gerais, FCMMG, Belo Horizonte, MG, Brazil.
| | - Wallace Camargos
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Priscila A C Valadão
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rubens S Garcias
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Hermann A Rodrigues
- Departamento de Ciências Básicas da Vida, Instituto de Ciências da Vida, Universidade Federal de Juiz de Fora, Campus Governador Valadares, UFJF, Governador Valadares, MG, Brazil
| | - Jéssica N Andrade
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vanessa P Teixeira
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lígia A Naves
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walter L G Cavalcante
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcia Gallaci
- Departamento de Farmacologia, Instituto de Biociências, UNESP, Distrito de Rubião Jr., Botucatu, São Paulo, Brazil
| | - Silvia Guatimosim
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vânia F Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell Biology, University of Western Ontario, London, ON, Canada
| | - Marco A M Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell Biology, University of Western Ontario, London, ON, Canada
| | - Cristina Guatimosim
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
22
|
Lamond A, Buckley D, O'Dea J, Turner L. Variants of SLC18A3 leading to congenital myasthenic syndrome in two children with varying presentations. BMJ Case Rep 2021; 14:14/1/e237799. [PMID: 33462016 PMCID: PMC7813295 DOI: 10.1136/bcr-2020-237799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
This report describes the variation in presentation of two unrelated patients found to have a rare form of presynaptic congenital myasthenic syndrome. Both patients presented with hypotonia, ptosis, poor weight gain and apneic episodes. Through whole exome sequencing, our patients were found to have the same likely pathogenic biallelic variants in W315X and I200N of SLC18A3, encoding vesicular acetylcholine transporter (VAChT). These specific variants in SLC18A3 have not been previously described in the literature. We illustrate the variety in clinical presentation and course of children with mutations in SLC18A3, leading to presynaptic congenital myasthenic syndrome through VAChT deficiency.
Collapse
Affiliation(s)
- Allison Lamond
- Pediatrics, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| | - David Buckley
- Pediatric Neurology, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| | - Jennifer O'Dea
- Pediatrics, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| | - Lesley Turner
- Genetics, Memorial University of Newfoundland Faculty of Medicine, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
23
|
Rodríguez Cruz PM, Hughes I, Manzur A, Munot P, Ramdas S, Wright R, Breen C, Pitt M, Pagnamenta AT, Taylor JC, Palace J, Beeson D. Presynaptic congenital myasthenic syndrome due to three novel mutations in SLC5A7 encoding the sodium-dependant high-affinity choline transporter. Neuromuscul Disord 2021; 31:21-28. [DOI: 10.1016/j.nmd.2020.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 01/14/2023]
|
24
|
Bhat S, El-Kasaby A, Freissmuth M, Sucic S. Functional and Biochemical Consequences of Disease Variants in Neurotransmitter Transporters: A Special Emphasis on Folding and Trafficking Deficits. Pharmacol Ther 2020; 222:107785. [PMID: 33310157 PMCID: PMC7612411 DOI: 10.1016/j.pharmthera.2020.107785] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023]
Abstract
Neurotransmitters, such as γ-aminobutyric acid, glutamate, acetyl choline, glycine and the monoamines, facilitate the crosstalk within the central nervous system. The designated neurotransmitter transporters (NTTs) both release and take up neurotransmitters to and from the synaptic cleft. NTT dysfunction can lead to severe pathophysiological consequences, e.g. epilepsy, intellectual disability, or Parkinson’s disease. Genetic point mutations in NTTs have recently been associated with the onset of various neurological disorders. Some of these mutations trigger folding defects in the NTT proteins. Correct folding is a prerequisite for the export of NTTs from the endoplasmic reticulum (ER) and the subsequent trafficking to their pertinent site of action, typically at the plasma membrane. Recent studies have uncovered some of the key features in the molecular machinery responsible for transporter protein folding, e.g., the role of heat shock proteins in fine-tuning the ER quality control mechanisms in cells. The therapeutic significance of understanding these events is apparent from the rising number of reports, which directly link different pathological conditions to NTT misfolding. For instance, folding-deficient variants of the human transporters for dopamine or GABA lead to infantile parkinsonism/dystonia and epilepsy, respectively. From a therapeutic point of view, some folding-deficient NTTs are amenable to functional rescue by small molecules, known as chemical and pharmacological chaperones.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
25
|
Cacabelos R. Pharmacogenetic considerations when prescribing cholinesterase inhibitors for the treatment of Alzheimer's disease. Expert Opin Drug Metab Toxicol 2020; 16:673-701. [PMID: 32520597 DOI: 10.1080/17425255.2020.1779700] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Cholinergic dysfunction, demonstrated in the late 1970s and early 1980s, led to the introduction of acetylcholinesterase inhibitors (AChEIs) in 1993 (Tacrine) to enhance cholinergic neurotransmission as the first line of treatment against Alzheimer's disease (AD). The new generation of AChEIs, represented by Donepezil (1996), Galantamine (2001) and Rivastigmine (2002), is the only treatment for AD to date, together with Memantine (2003). AChEIs are not devoid of side-effects and their cost-effectiveness is limited. An option to optimize the correct use of AChEIs is the implementation of pharmacogenetics (PGx) in the clinical practice. AREAS COVERED (i) The cholinergic system in AD, (ii) principles of AD PGx, (iii) PGx of Donepezil, Galantamine, Rivastigmine, Huperzine and other treatments, and (iv) practical recommendations. EXPERT OPINION The most relevant genes influencing AChEI efficacy and safety are APOE and CYPs. APOE-4 carriers are the worst responders to AChEIs. With the exception of Rivastigmine (UGT2B7, BCHE-K), the other AChEIs are primarily metabolized via CYP2D6, CYP3A4, and UGT enzymes, with involvement of ABC transporters and cholinergic genes (CHAT, ACHE, BCHE, SLC5A7, SLC18A3, CHRNA7) in most ethnic groups. Defective variants may affect the clinical response to AChEIs. PGx geno-phenotyping is highly recommended prior to treatment.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine , Bergondo, Corunna, Spain
| |
Collapse
|
26
|
Xiao T, Wu LW. [Advances in the diagnosis and treatment of congenital myasthenic syndrome]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:672-676. [PMID: 32571471 PMCID: PMC7390217 DOI: 10.7499/j.issn.1008-8830.1912095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/24/2020] [Indexed: 06/11/2023]
Abstract
Congenital myasthenic syndrome (CMS) is a group of clinical and genetic heterogeneous diseases caused by impaired neuromuscular transmission due to genetic defects. At present, it has been reported that more than 30 genes can cause CMS. All CMS subtypes have the clinical features of fatigue and muscle weakness, but age of onset, symptoms, and treatment response vary with the molecular mechanisms underlying genetic defects. Pharmacotherapy and symptomatic/supportive treatment are the main methods for the treatment of CMS, and antisense oligonucleotide technology has been proven to be beneficial for CHRNA 1-related CMS in animals. Since CMS is a group of increasingly recognized clinical and genetic heterogeneous diseases, an understanding of the latest knowledge and research advances in its clinical features, genetic research, and treatment helps to give early diagnosis and treatment as well as gain a deeper understanding of the pathogenesis of CMS, so as to make new breakthroughs in the treatment of CMS.
Collapse
Affiliation(s)
- Ting Xiao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China.
| | | |
Collapse
|
27
|
Xiao T, Wu LW. [Advances in the diagnosis and treatment of congenital myasthenic syndrome]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:672-676. [PMID: 32571471 PMCID: PMC7390217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/24/2020] [Indexed: 11/12/2023]
Abstract
Congenital myasthenic syndrome (CMS) is a group of clinical and genetic heterogeneous diseases caused by impaired neuromuscular transmission due to genetic defects. At present, it has been reported that more than 30 genes can cause CMS. All CMS subtypes have the clinical features of fatigue and muscle weakness, but age of onset, symptoms, and treatment response vary with the molecular mechanisms underlying genetic defects. Pharmacotherapy and symptomatic/supportive treatment are the main methods for the treatment of CMS, and antisense oligonucleotide technology has been proven to be beneficial for CHRNA 1-related CMS in animals. Since CMS is a group of increasingly recognized clinical and genetic heterogeneous diseases, an understanding of the latest knowledge and research advances in its clinical features, genetic research, and treatment helps to give early diagnosis and treatment as well as gain a deeper understanding of the pathogenesis of CMS, so as to make new breakthroughs in the treatment of CMS.
Collapse
Affiliation(s)
- Ting Xiao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China.
| | | |
Collapse
|
28
|
The congenital myasthenic syndromes: expanding genetic and phenotypic spectrums and refining treatment strategies. Curr Opin Neurol 2020; 32:696-703. [PMID: 31361628 PMCID: PMC6735524 DOI: 10.1097/wco.0000000000000736] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Congenital myasthenic syndromes (CMS) are a group of heterogeneous inherited disorders caused by mutations in genes encoding proteins whose function is essential for the integrity of neuromuscular transmission. This review updates the reader on the expanding phenotypic spectrum and suggested improved treatment strategies.
Collapse
|
29
|
Microbiota Alters Urinary Bladder Weight and Gene Expression. Microorganisms 2020; 8:microorganisms8030421. [PMID: 32192034 PMCID: PMC7143536 DOI: 10.3390/microorganisms8030421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
We studied the effect of microbiota on the transcriptome and weight of the urinary bladder by comparing germ-free (GF) and specific pathogen-free (SPF) housed mice. In total, 97 genes were differently expressed (fold change > ±2; false discovery rate (FDR) p-value < 0.01) between the groups, including genes regulating circadian rhythm (Per1, Per2 and Per3), extracellular matrix (Spo1, Spon2), and neuromuscular synaptic transmission (Slc18a3, Slc5a7, Chrnb4, Chrna3, Snap25). The highest increase in expression was observed for immunoglobulin genes (Igkv1-122, Igkv4-68) of unknown function, but surprisingly the absence of microbiota did not change the expression of the genes responsible for recognizing microbes and their products. We found that urinary bladder weight was approximately 25% lighter in GF mice (p = 0.09 for males, p = 0.005 for females) and in mice treated with broad spectrum of antibiotics (p = 0.0002). In conclusion, our data indicate that microbiota is an important determinant of urinary bladder physiology controlling its gene expression and size.
Collapse
|
30
|
Balaraju S, Töpf A, McMacken G, Kumar VP, Pechmann A, Roper H, Vengalil S, Polavarapu K, Nashi S, Mahajan NP, Barbosa IA, Deshpande C, Taylor RW, Cossins J, Beeson D, Laurie S, Kirschner J, Horvath R, McFarland R, Nalini A, Lochmüller H. Congenital myasthenic syndrome with mild intellectual disability caused by a recurrent SLC25A1 variant. Eur J Hum Genet 2020; 28:373-377. [PMID: 31527857 PMCID: PMC7029005 DOI: 10.1038/s41431-019-0506-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 11/10/2022] Open
Abstract
Congenital myasthenic syndromes (CMS) are a clinically and genetically heterogeneous group of disorders caused by mutations which lead to impaired neuromuscular transmission. SLC25A1 encodes a mitochondrial citrate carrier, associated mainly with the severe neurometabolic disease combined D-2- and L-2-hydroxyglutaric aciduria (D/L-2-HGA). We previously reported a single family with a homozygous missense variant in SLC25A1 with a phenotype restricted to relatively mild CMS with intellectual disability, but to date no additional cases of this CMS subtype had been reported. Here, we performed whole exome sequencing (WES) in three additional and unrelated families presenting with CMS and mild intellectual disability to identify the underlying causative gene. The WES analysis revealed the presence of a homozygous c.740G>A; p.(Arg247Gln) missense SLC25A1 variant, the same SLC25A1 variant as identified in the original family with this phenotype. Electron microscopy of muscle from two cases revealed enlarged and accumulated mitochondria. Haplotype analysis performed in two unrelated families suggested that this variant is a result of recurrent mutation and not a founder effect. This suggests that p.(Arg247Gln) is associated with a relatively mild CMS phenotype with subtle mitochondrial abnormalities, while other variants in this gene cause more severe neurometabolic disease. In conclusion, the p.(Arg247Gln) SLC25A1 variant should be considered in patients presenting with a presynaptic CMS phenotype, particularly with accompanying intellectual disability.
Collapse
Affiliation(s)
- Sunitha Balaraju
- Department of Clinical Neurosciences, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Ana Töpf
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Grace McMacken
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Veeramani Preethish Kumar
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
- Department of Clinical Neurosciences, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Astrid Pechmann
- Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Helen Roper
- Department of Paediatrics, Heartlands Hospital, Birmingham, UK
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Kiran Polavarapu
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
- Department of Clinical Neurosciences, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Niranjan Prakash Mahajan
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Ines A Barbosa
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences King's College London, London, UK
| | - Charu Deshpande
- Clinical Genetics Unit, Guys and St Thomas' NHS Foundation Trust, London, UK
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Judith Cossins
- Neurosciences Group, Nuffield Department of Clinical Neuroscience, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - David Beeson
- Neurosciences Group, Nuffield Department of Clinical Neuroscience, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Steven Laurie
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India.
| | - Hanns Lochmüller
- 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.
- Children's Hospital of Eastern Ontario Research Institute; Division of Neurology, Department of Medicine, The Ottawa Hospital; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
31
|
Leite Schetino LP, Fonseca M, Magalhães Gomes MPS, Costa Valadão PA, Camargo WL, Rodrigues HA, Andrade JN, Arantes‐Costa FM, Naves LA, Prado CM, Prado VF, Prado MAM, Guatimosim C. Evaluation of the neuromuscular junction in a middle‐aged mouse model of congenital myasthenic syndrome. Muscle Nerve 2019; 60:790-800. [DOI: 10.1002/mus.26710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022]
Affiliation(s)
| | - Matheus Fonseca
- Laboratório Nacional de BiociênciasCentro Nacional de Pesquisa em Energia e Materiais Campinas São Paulo Brazil
| | | | | | - Wallace Lucio Camargo
- Departamento de Fisiologia e BiofísicaUniversidade Federal de Minas Gerais Belo Horizonte Minas Gerais Brazil
| | - Hermann Alecsandro Rodrigues
- Departamento de Ciências Básicas da Vida, Instituto de Ciências da VidaUniversidade Federal de Juiz de Fora Campus Governador Valadares Minas Gerais Brazil
| | - Jéssica Neves Andrade
- Departamento de MorfologiaUniversidade Federal de Minas Gerais Belo Horizonte Minas Gerais Brazil
| | | | - Lígia Araujo Naves
- Departamento de Fisiologia e BiofísicaUniversidade Federal de Minas Gerais Belo Horizonte Minas Gerais Brazil
| | - Carla Máximo Prado
- Departmento de BiociênciasUniversidade Federal de São Paulo, Campus Baixada Santista São Paulo Brazil
| | - Vânia Ferreira Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell BiologyUniversity of Western Ontario London Ontario Canada
| | - Marco Antônio Máximo Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell BiologyUniversity of Western Ontario London Ontario Canada
| | - Cristina Guatimosim
- Departamento de MorfologiaUniversidade Federal de Minas Gerais Belo Horizonte Minas Gerais Brazil
| |
Collapse
|
32
|
Nicole S, Azuma Y, Bauché S, Eymard B, Lochmüller H, Slater C. Congenital Myasthenic Syndromes or Inherited Disorders of Neuromuscular Transmission: Recent Discoveries and Open Questions. J Neuromuscul Dis 2019; 4:269-284. [PMID: 29125502 PMCID: PMC5701762 DOI: 10.3233/jnd-170257] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Congenital myasthenic syndromes (CMS) form a heterogeneous group of rare diseases characterized by fatigable muscle weakness. They are genetically-inherited and caused by defective synaptic transmission at the cholinergic neuromuscular junction (NMJ). The number of genes known to cause CMS when mutated is currently 30, and the relationship between fatigable muscle weakness and defective functions is quite well-understood for many of them. However, some of the most recent discoveries in individuals with CMS challenge our knowledge of the NMJ, where the basis of the pathology has mostly been investigated in animal models. Frontier forms between CMS and congenital myopathy, which have been genetically and clinically identified, underline the poorly understood interplay between the synaptic and extrasynaptic molecules in the neuromuscular system. In addition, precise electrophysiological and histopathological investigations of individuals with CMS suggest an important role of NMJ plasticity in the response to CMS pathogenesis. While efficient drug-based treatments are already available to improve neuromuscular transmission for most forms of CMS, others, as well as neurological and muscular comorbidities, remain resistant. Taken together, the available pathological data point to physiological issues which remain to be understood in order to achieve precision medicine with efficient therapeutics for all individuals suffering from CMS.
Collapse
Affiliation(s)
- Sophie Nicole
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France
| | - Yoshiteru Azuma
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Stéphanie Bauché
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France
| | - Bruno Eymard
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, 75013 Paris, France
- AP-HP, Hôpital Pitié-Salpétrière, 75013 Paris, France
| | - Hanns Lochmüller
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Clarke Slater
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| |
Collapse
|
33
|
Hakonen AH, Polvi A, Saloranta C, Paetau A, Heikkilä P, Almusa H, Ellonen P, Jakkula E, Saarela J, Aittomäki K. SLC18A3 variants lead to fetal akinesia deformation sequence early in pregnancy. Am J Med Genet A 2019; 179:1362-1365. [PMID: 31059209 DOI: 10.1002/ajmg.a.61186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/01/2019] [Accepted: 04/23/2019] [Indexed: 11/10/2022]
Abstract
Fetal akinesia deformation sequence (FADS) and lethal multiple pterygium syndrome (LMPS) are clinically overlapping syndromes manifesting with reduced or absent fetal movement, arthrogryposis, and several anomalies during fetal life. The etiology of these syndromes is heterogeneous, and in many cases it remains unknown. In order to determine the genetic etiology of FADS in two fetuses with fetal akinesia, arthrogryposis, edema, and partial cleft palate, we utilized exome sequencing. Our investigations revealed a homozygous nonsense variant [c.1116C>A, p.(Cys372Ter)] in the SLC18A3 gene, which encodes for the vesicular acetylcholine transporter (VAChT) responsible for active transport of acetylcholine in the neuromuscular junction. This is the first description of a nonsense variant in the SLC18A3 gene, as only missense variants and whole gene deletions have been previously identified in patients. The previously detected SLC18A3 defects have been associated with congenital myasthenic syndromes, and therefore our findings extend the clinical spectrum of SLC18A3 defects to severe prenatal phenotypes. Our findings suggest that nonsense variants in SLC18A3 cause a more severe phenotype than missense variants and are in line with previous studies showing a lethal phenotype in VAChT knockout mice. Our results underline the importance of including SLC18A3 sequencing in the differential diagnostics of fetuses with arthrogryposis, FADS, or LMPS of unknown etiology.
Collapse
Affiliation(s)
- Anna H Hakonen
- Department of Clinical Genetics, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anne Polvi
- Department of Clinical Genetics, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Carola Saloranta
- Department of Clinical Genetics, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi Heikkilä
- Department of Pathology, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Eveliina Jakkula
- Department of Clinical Genetics, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Janna Saarela
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Kristiina Aittomäki
- Department of Clinical Genetics, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
34
|
Nicolau S, Milone M. The Electrophysiology of Presynaptic Congenital Myasthenic Syndromes With and Without Facilitation: From Electrodiagnostic Findings to Molecular Mechanisms. Front Neurol 2019; 10:257. [PMID: 30941097 PMCID: PMC6433874 DOI: 10.3389/fneur.2019.00257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/26/2019] [Indexed: 11/13/2022] Open
Abstract
Congenital myasthenic syndromes (CMS) are a group of inherited disorders of neuromuscular transmission most commonly presenting with early onset fatigable weakness, ptosis, and ophthalmoparesis. CMS are classified according to the localization of the causative molecular defect. CMS with presynaptic dysfunction can be caused by mutations in several different genes, including those involved in acetylcholine synthesis, its packaging into synaptic vesicles, vesicle docking, and release from the presynaptic nerve terminal and neuromuscular junction development and maintenance. Electrodiagnostic testing is key in distinguishing CMS from other neuromuscular disorders with similar clinical features as well as for revealing features pointing to a specific molecular diagnosis. A decremental response on low-frequency repetitive nerve stimulation (RNS) is present in most presynaptic CMS. In CMS with deficits in acetylcholine resynthesis however, a decrement may only appear after conditioning with exercise or high-frequency RNS and characteristically displays a slow recovery. Facilitation occurs in CMS caused by mutations in VAMP1, UNC13A, SYT2, AGRN, LAMA5. By contrast, facilitation is absent in the other presynaptic CMS described to date. An understanding of the underlying molecular mechanisms therefore assists the interpretation of electrodiagnostic findings in patients with suspected CMS.
Collapse
Affiliation(s)
- Stefan Nicolau
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | | |
Collapse
|
35
|
Abstract
OBJECTIVES Congenital myasthenic syndromes (CMSs) are a genotypically and phenotypically heterogeneous group of neuromuscular disorders, which have in common an impaired neuromuscular transmission. Since the field of CMSs is steadily expanding, the present review aimed at summarizing and discussing current knowledge and recent advances concerning the etiology, clinical presentation, diagnosis, and treatment of CMSs. METHODS Systematic literature review. RESULTS Currently, mutations in 32 genes are made responsible for autosomal dominant or autosomal recessive CMSs. These mutations concern 8 presynaptic, 4 synaptic, 15 post-synaptic, and 5 glycosilation proteins. These proteins function as ion-channels, enzymes, or structural, signalling, sensor, or transporter proteins. The most common causative genes are CHAT, COLQ, RAPSN, CHRNE, DOK7, and GFPT1. Phenotypically, these mutations manifest as abnormal fatigability or permanent or fluctuating weakness of extra-ocular, facial, bulbar, axial, respiratory, or limb muscles, hypotonia, or developmental delay. Cognitive disability, dysmorphism, neuropathy, or epilepsy are rare. Low- or high-frequency repetitive nerve stimulation may show an abnormal increment or decrement, and SF-EMG an increased jitter or blockings. Most CMSs respond favourably to acetylcholine-esterase inhibitors, 3,4-diamino-pyridine, salbutamol, albuterol, ephedrine, fluoxetine, or atracurium. CONCLUSIONS CMSs are an increasingly recognised group of genetically transmitted defects, which usually respond favorably to drugs enhancing the neuromuscular transmission. CMSs need to be differentiated from neuromuscular disorders due to muscle or nerve dysfunction.
Collapse
Affiliation(s)
- Josef Finsterer
- Krankenanstalt Rudolfstiftung, Messerli Institute, Veterinary University of Vienna, Postfach 20, 1180, Vienna, Austria.
| |
Collapse
|
36
|
Magalhães-Gomes MPS, Motta-Santos D, Schetino LPL, Andrade JN, Bastos CP, Guimarães DAS, Vaughan SK, Martinelli PM, Guatimosim S, Pereira GS, Coimbra CC, Prado VF, Prado MAM, Valdez G, Guatimosim C. Fast and slow-twitching muscles are differentially affected by reduced cholinergic transmission in mice deficient for VAChT: A mouse model for congenital myasthenia. Neurochem Int 2018; 120:1-12. [PMID: 30003945 PMCID: PMC6421860 DOI: 10.1016/j.neuint.2018.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/27/2018] [Accepted: 07/07/2018] [Indexed: 12/15/2022]
Abstract
Congenital myasthenic syndromes (CMS) result from reduced cholinergic transmission at neuromuscular junctions (NMJs). While the etiology of CMS varies, the disease is characterized by muscle weakness. To date, it remains unknown if CMS causes long-term and irreversible changes to skeletal muscles. In this study, we examined skeletal muscles in a mouse line with reduced expression of Vesicular Acetylcholine Transporter (VAChT, mouse line herein called VAChT-KDHOM). We examined this mouse line for several reasons. First, VAChT plays a central function in loading acetylcholine (ACh) into synaptic vesicles and releasing it at NMJs, in addition to other cholinergic nerve endings. Second, loss of function mutations in VAChT causes myasthenia in humans. Importantly, VAChT-KDHOM present with reduced ACh and muscle weakness, resembling CMS. We evaluated the morphology, fiber type (myosin heavy chain isoforms), and expression of muscle-related genes in the extensor digitorum longus (EDL) and soleus muscles. This analysis revealed that while muscle fibers atrophy in the EDL, they hypertrophy in the soleus muscle of VAChT-KDHOM mice. Along with these cellular changes, skeletal muscles exhibit altered levels of markers for myogenesis (Pax-7, Myogenin, and MyoD), oxidative metabolism (PGC1-α and MTND1), and protein degradation (Atrogin1 and MuRF1) in VAChT-KDHOM mice. Importantly, we demonstrate that deleterious changes in skeletal muscles and motor deficits can be partially reversed following the administration of the cholinesterase inhibitor, pyridostigmine in VAChT-KDHOM mice. These findings reveal that fast and slow type muscles differentially respond to cholinergic deficits. Additionally, this study shows that the adverse effects of cholinergic transmission, as in the case of CMS, on fast and slow type skeletal muscles are reversible.
Collapse
Affiliation(s)
| | - Daisy Motta-Santos
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Departamento de Esportes, EEFFTO, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luana P L Schetino
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jéssica N Andrade
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cristiane P Bastos
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Sydney K Vaughan
- Virginia Tech Carilion Research Institute, Roanoke, VA, USA; Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Patrícia M Martinelli
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Silvia Guatimosim
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Grace S Pereira
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Candido C Coimbra
- Departamento de Fisiologia e Biofísica, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vânia F Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell Biology, University of Western Ontario, London, ON, Canada
| | - Marco A M Prado
- Robarts Research Institute and Department of Physiology and Pharmacology and Anatomy & Cell Biology, University of Western Ontario, London, ON, Canada
| | - Gregorio Valdez
- Virginia Tech Carilion Research Institute, Roanoke, VA, USA; Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Cristina Guatimosim
- Departamento de Morfologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
37
|
Farmakidis C, Pasnoor M, Barohn RJ, Dimachkie MM. Congenital Myasthenic Syndromes: a Clinical and Treatment Approach. Curr Treat Options Neurol 2018; 20:36. [DOI: 10.1007/s11940-018-0520-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
38
|
Abstract
PURPOSE OF REVIEW Summarize features of the currently recognized congenital myasthenic syndromes (CMS) with emphasis on novel findings identified in the past 6 years. RECENT FINDINGS Since the last review of the CMS in this journal in 2012, several novel CMS were identified. The identified disease proteins are SNAP25B, synaptotagmin 2, Munc13-1, synaptobrevin-1, GFPT1, DPAGT1, ALG2, ALG14, Agrin, GMPPB, LRP4, myosin 9A, collagen 13A1, the mitochondrial citrate carrier, PREPL, LAMA5, the vesicular ACh transporter, and the high-affinity presynaptic choline transporter. Exome sequencing has provided a powerful tool for identifying novel CMS. Identifying the disease genes is essential for determining optimal therapy. The landscape of the CMS is still unfolding.
Collapse
Affiliation(s)
- Andrew G Engel
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
39
|
Rodríguez Cruz PM, Palace J, Beeson D. The Neuromuscular Junction and Wide Heterogeneity of Congenital Myasthenic Syndromes. Int J Mol Sci 2018; 19:ijms19061677. [PMID: 29874875 PMCID: PMC6032286 DOI: 10.3390/ijms19061677] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/17/2018] [Accepted: 05/21/2018] [Indexed: 01/16/2023] Open
Abstract
Congenital myasthenic syndromes (CMS) are genetic disorders characterised by impaired neuromuscular transmission. This review provides an overview on CMS and highlights recent advances in the field, including novel CMS causative genes and improved therapeutic strategies. CMS due to mutations in SLC5A7 and SLC18A3, impairing the synthesis and recycling of acetylcholine, have recently been described. In addition, a novel group of CMS due to mutations in SNAP25B, SYT2, VAMP1, and UNC13A1 encoding molecules implicated in synaptic vesicles exocytosis has been characterised. The increasing number of presynaptic CMS exhibiting CNS manifestations along with neuromuscular weakness demonstrate that the myasthenia can be only a small part of a much more extensive disease phenotype. Moreover, the spectrum of glycosylation abnormalities has been increased with the report that GMPPB mutations can cause CMS, thus bridging myasthenic disorders with dystroglycanopathies. Finally, the discovery of COL13A1 mutations and laminin α5 deficiency has helped to draw attention to the role of extracellular matrix proteins for the formation and maintenance of muscle endplates. The benefit of β2-adrenergic agonists alone or combined with pyridostigmine or 3,4-Dyaminopiridine is increasingly being reported for different subtypes of CMS including AChR-deficiency and glycosylation abnormalities, thus expanding the therapeutic repertoire available.
Collapse
Affiliation(s)
- Pedro M Rodríguez Cruz
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford OX3 9DS, UK.
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford OX3 9DS, UK.
| |
Collapse
|
40
|
Beeson D, Cossins J, Rodriguez-Cruz P, Maxwell S, Liu WW, Palace J. Myasthenic syndromes due to defects in COL13A1 and in the N-linked glycosylation pathway. Ann N Y Acad Sci 2018; 1413:163-169. [DOI: 10.1111/nyas.13576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Affiliation(s)
- David Beeson
- Neurosciences Group, Nuffield Department of Clinical Neuroscience; Weatherall Institute of Molecular Medicine; The John Radcliffe Oxford UK
| | - Judith Cossins
- Neurosciences Group, Nuffield Department of Clinical Neuroscience; Weatherall Institute of Molecular Medicine; The John Radcliffe Oxford UK
| | - Pedro Rodriguez-Cruz
- Neurosciences Group, Nuffield Department of Clinical Neuroscience; Weatherall Institute of Molecular Medicine; The John Radcliffe Oxford UK
| | - Susan Maxwell
- Neurosciences Group, Nuffield Department of Clinical Neuroscience; Weatherall Institute of Molecular Medicine; The John Radcliffe Oxford UK
| | - Wei-Wei Liu
- Neurosciences Group, Nuffield Department of Clinical Neuroscience; Weatherall Institute of Molecular Medicine; The John Radcliffe Oxford UK
| | - Jacqueline Palace
- Nuffield Department of Clinical Neuroscience; Level 3 The West Wing; The John Radcliffe Oxford UK
| |
Collapse
|
41
|
McMacken G, Whittaker RG, Evangelista T, Abicht A, Dusl M, Lochmüller H. Congenital myasthenic syndrome with episodic apnoea: clinical, neurophysiological and genetic features in the long-term follow-up of 19 patients. J Neurol 2018; 265:194-203. [PMID: 29189923 PMCID: PMC5760613 DOI: 10.1007/s00415-017-8689-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/19/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Congenital myasthenic syndrome with episodic apnoea (CMS-EA) is a rare but potentially treatable cause of apparent life-threatening events in infancy. The underlying mechanisms for sudden and recurrent episodes of respiratory arrest in these patients are unclear. Whilst CMS-EA is most commonly caused by mutations in CHAT, the list of associated genotypes is expanding. METHODS We reviewed clinical information from 19 patients with CMS-EA, including patients with mutations in CHAT, SLC5A7 and RAPSN, and patients lacking a genetic diagnosis. RESULTS Lack of genetic diagnosis was more common in CMS-EA than in CMS without EA (56% n = 18, compared to 7% n = 97). Most patients manifested intermittent apnoea in the first 4 months of life (74%, n = 14). A degree of clinical improvement with medication was observed in most patients (74%, n = 14), but the majority of cases also showed a tendency towards complete remission of apnoeic events with age (mean age of resolution 2 years 5 months). Signs of impaired neuromuscular transmission were detected on neurophysiology studies in 79% (n = 15) of cases, but in six cases, this was only apparent following specific neurophysiological testing protocols (prolonged high-frequency stimulation). CONCLUSIONS A relatively large proportion of CMS-EA remains genetically undiagnosed, which suggests the existence of novel causative CMS genes which remain uncharacterised. In light of the potential for recurrent life-threatening apnoeas in early life and the positive response to therapy, early diagnostic consideration of CMS-EA is critical, but without specific neurophysiology tests, it may go overlooked.
Collapse
Affiliation(s)
- Grace McMacken
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK.
| | - Roger G Whittaker
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Teresinha Evangelista
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Angela Abicht
- Friedrich-Baur-Institute, Ludwig Maximilians University, Munich, Germany
| | - Marina Dusl
- Friedrich-Baur-Institute, Ludwig Maximilians University, Munich, Germany
| | - Hanns Lochmüller
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
42
|
Schwartz M, Sternberg D, Whalen S, Afenjar A, Isapof A, Chabrol B, Portnoï MF, Heide S, Keren B, Chantot-Bastaraud S, Siffroi JP. How chromosomal deletions can unmask recessive mutations? Deletions in 10q11.2 associated with CHAT or SLC18A3 mutations lead to congenital myasthenic syndrome. Am J Med Genet A 2017; 176:151-155. [PMID: 29130637 DOI: 10.1002/ajmg.a.38515] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 11/06/2022]
Abstract
A congenital myasthenia was suspected in two unrelated children with very similar phenotypes including several episodes of severe dyspnea. Both children had a 10q11.2 deletion revealed by Single Nucleotide Polymorphisms array or by Next Generation Sequencing analysis. The deletion was inherited from the healthy mother in the first case. These deletions unmasked a recessive mutation at the same locus in both cases, but in two different genes: CHAT and SLC18A3.
Collapse
Affiliation(s)
- Mathias Schwartz
- AP-HP, Département de Génétique Médicale, Hôpital Armand Trousseau, UPMC, Paris, France
| | - Damien Sternberg
- AP-HP, Hôpital Pitié-Salpêtrière, Biochimie et Génétique, Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, ICM, Paris, France
| | - Sandra Whalen
- AP-HP, Hôpital Armand Trousseau, UF de Génétique Clinique, UPMC, Paris, France
| | - Alexandra Afenjar
- AP-HP, Hôpital Armand Trousseau, UF de Génétique Clinique, UPMC, Paris, France
| | - Arnaud Isapof
- AP-HP, Hôpital Armand Trousseau, Unité de Neuropédiatrie et Pathologie du Développement, UPMC, Paris, France
| | - Brigitte Chabrol
- CHU de Marseille, Hôpital de la Timone, Service de Neurologie Pédiatrique, Inserm U1127, CNRS UMR 7225, UPMC, Centre de Recherche de l'Institut du Cerveau et de la Moëlle épinière, Paris, France
| | - Marie-France Portnoï
- AP-HP, Département de Génétique Médicale, Hôpital Armand Trousseau, UPMC, Paris, France
| | - Solveig Heide
- AP-HP, Département de Génétique Médicale, Hôpital Armand Trousseau, UPMC, Paris, France
| | - Boris Keren
- AP-HP, Département de Génétique, Hôpital de la Pitié Salpêtrière, Paris, France
| | | | - Jean-Pierre Siffroi
- AP-HP, Département de Génétique Médicale, Hôpital Armand Trousseau, UPMC, Paris, France
| |
Collapse
|
43
|
Mutations in GFPT1-related congenital myasthenic syndromes are associated with synaptic morphological defects and underlie a tubular aggregate myopathy with synaptopathy. J Neurol 2017; 264:1791-1803. [DOI: 10.1007/s00415-017-8569-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/03/2017] [Accepted: 07/11/2017] [Indexed: 12/22/2022]
|
44
|
Janickova H, Prado VF, Prado MAM, El Mestikawy S, Bernard V. Vesicular acetylcholine transporter (VAChT) over-expression induces major modifications of striatal cholinergic interneuron morphology and function. J Neurochem 2017. [DOI: 10.1111/jnc.14105] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Helena Janickova
- Department of Physiology and Pharmacology and Department of Anatomy & Cell Biology; Robarts Research Institute; Molecular Medicine Laboratories; The University of Western Ontario; London Ontario Canada
| | - Vania F. Prado
- Department of Physiology and Pharmacology and Department of Anatomy & Cell Biology; Robarts Research Institute; Molecular Medicine Laboratories; The University of Western Ontario; London Ontario Canada
| | - Marco A. M. Prado
- Department of Physiology and Pharmacology and Department of Anatomy & Cell Biology; Robarts Research Institute; Molecular Medicine Laboratories; The University of Western Ontario; London Ontario Canada
| | - Salah El Mestikawy
- Sorbonne Universités; Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130; Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS); Paris France
- Department of Psychiatry; Douglas Mental Health University Institute; McGill University; Montreal Canada
| | - Véronique Bernard
- Sorbonne Universités; Université Pierre et Marie Curie UM 119 - CNRS UMR 8246 - INSERM U1130; Neurosciences Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS); Paris France
| |
Collapse
|
45
|
Salpietro V, Lin W, Vedove AD, Storbeck M, Liu Y, Efthymiou S, Manole A, Wiethoff S, Ye Q, Saggar A, McElreavey K, Krishnakumar SS, Pitt M, Bello OD, Rothman JE, Basel‐Vanagaite L, Hubshman MW, Aharoni S, Manzur AY, Wirth B, Houlden H. Homozygous mutations in VAMP1 cause a presynaptic congenital myasthenic syndrome. Ann Neurol 2017; 81:597-603. [PMID: 28253535 PMCID: PMC5413866 DOI: 10.1002/ana.24905] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/24/2017] [Accepted: 02/24/2017] [Indexed: 11/07/2022]
Abstract
We report 2 families with undiagnosed recessive presynaptic congenital myasthenic syndrome (CMS). Whole exome or genome sequencing identified segregating homozygous variants in VAMP1: c.51_64delAGGTGGGGGTCCCC in a Kuwaiti family and c.146G>C in an Israeli family. VAMP1 is crucial for vesicle fusion at presynaptic neuromuscular junction (NMJ). Electrodiagnostic examination showed severely low compound muscle action potentials and presynaptic impairment. We assessed the effect of the nonsense mutation on mRNA levels and evaluated the NMJ transmission in VAMP1lew/lew mice, observing neurophysiological features of presynaptic impairment, similar to the patients. Taken together, our findings highlight VAMP1 homozygous mutations as a cause of presynaptic CMS. Ann Neurol 2017;81:597–603
Collapse
Affiliation(s)
- Vincenzo Salpietro
- Department of Molecular Neuroscience, Institute of NeurologyUniversity College London Institute of NeurologyLondonUnited Kingdom
| | - Weichun Lin
- Department of NeuroscienceUniversity of Texas Southwestern Medical CenterDallasTX
| | - Andrea Delle Vedove
- Institute of Human Genetics, Center for Molecular Medicine CologneCologneGermany
- Institute for GeneticsUniversity of CologneCologneGermany
| | - Markus Storbeck
- Institute of Human Genetics, Center for Molecular Medicine CologneCologneGermany
- Institute for GeneticsUniversity of CologneCologneGermany
| | - Yun Liu
- Department of NeuroscienceUniversity of Texas Southwestern Medical CenterDallasTX
| | - Stephanie Efthymiou
- Department of Molecular Neuroscience, Institute of NeurologyUniversity College London Institute of NeurologyLondonUnited Kingdom
| | - Andreea Manole
- Department of Molecular Neuroscience, Institute of NeurologyUniversity College London Institute of NeurologyLondonUnited Kingdom
| | - Sarah Wiethoff
- Department of Molecular Neuroscience, Institute of NeurologyUniversity College London Institute of NeurologyLondonUnited Kingdom
| | - Qiaohong Ye
- Department of NeuroscienceUniversity of Texas Southwestern Medical CenterDallasTX
| | - Anand Saggar
- St George's Hospital, National Health Service Foundation TrustLondonUnited Kingdom
| | | | - Shyam S. Krishnakumar
- Department of Cell BiologyYale School of MedicineNew HavenCT
- Department of Clinical and Experimental EpilepsyUniversity College London Institute of NeurologyLondonUnited Kingdom
| | | | - Matthew Pitt
- Department of Clinical NeurophysiologyGreat Ormond Street Hospital for Children, National Health Service Foundation TrustLondonUnited Kingdom
| | - Oscar D. Bello
- Department of Cell BiologyYale School of MedicineNew HavenCT
- Department of Clinical and Experimental EpilepsyUniversity College London Institute of NeurologyLondonUnited Kingdom
| | - James E. Rothman
- Department of Cell BiologyYale School of MedicineNew HavenCT
- Department of Clinical and Experimental EpilepsyUniversity College London Institute of NeurologyLondonUnited Kingdom
| | - Lina Basel‐Vanagaite
- Pediatric Genetics Unit, Schneider Children's Medical Center of IsraelPetach TikvaIsrael
- Raphael Recanati Genetic Institute, Rabin Medical CenterPetach TikvaIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Monika Weisz Hubshman
- Pediatric Genetics Unit, Schneider Children's Medical Center of IsraelPetach TikvaIsrael
- Raphael Recanati Genetic Institute, Rabin Medical CenterPetach TikvaIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Sharon Aharoni
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Institute of Child Neurology, Schneider Children's Medical Center of IsraelPetach TikvaIsrael
| | - Adnan Y. Manzur
- Department of Pediatric NeurologyDubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children National Health Service Foundation TrustLondonUnited Kingdom
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine CologneCologneGermany
| | - Henry Houlden
- Department of Molecular Neuroscience, Institute of NeurologyUniversity College London Institute of NeurologyLondonUnited Kingdom
| |
Collapse
|
46
|
Ardissone A, Moroni I, Bernasconi P, Brugnoni R. Congenital myasthenic syndrome: phenotypic variability in patients harbouring p.T159P mutation in CHRNE gene. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2017; 36:28-32. [PMID: 28690392 PMCID: PMC5479107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Congenital myasthenic syndromes (CMS) are rare and heterogeneous genetic diseases characterized by compromised neuromuscular transmission and clinical features of fatigable weakness; age at onset, presenting symptoms, distribution of weakness, and response to treatment differ depending on the underlying molecular defect. Mutations in one of the multiple genes, encoding proteins expressed at the neuromuscular junction, are currently known to be associated with subtypes of CMS. The most common CMS syndrome identified is associated with mutation in the CHRNE gene, causing principally muscle nicotinic acetylcholine receptor deficiency, that results in reduced receptor density on the postsynaptic membrane. We describe the clinical, neurophysiological and molecular features of two unrelated CMS Italian families with marked phenotypic variability, carrying the already reported p.T159P mutation in the CHRNE gene. Our report highlights clinical heterogeneity, intrafamily variability in spite of the same genotype and a possible gender effect; it confirms the efficacy and safety of salbutamol in patients who harbor mutations in the epsilon subunit of acetylcholine receptor.
Collapse
Affiliation(s)
- Anna Ardissone
- Child Neurology Unit, Foundation IRCCS Neurological Institute "Carlo Besta, Milan, Italy
| | - Isabella Moroni
- Child Neurology Unit, Foundation IRCCS Neurological Institute "Carlo Besta, Milan, Italy
| | - Pia Bernasconi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| | - Raffaella Brugnoni
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy,Address for correspondence: Dr Raffaella Brugnoni, Neurology IV, Neuroimmunology and Neuromuscular Diseases Unit, Foundation IRCCS Neurological Institute "Carlo Besta", via Celoria 11, 20133 Milan, Italy. Tel. +39 02 23944652. Fax +39 02 70633874. E-mail:
| |
Collapse
|
47
|
What's in the Literature? J Clin Neuromuscul Dis 2017; 18:165-175. [PMID: 28221312 DOI: 10.1097/cnd.0000000000000168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In this edition of this column, we review new studies concerning the pathophysiology, treatment, and outcomes of patients with necrotizing myopathy, genetic testing in congenital myopathies, and limb girdle muscular dystrophies, and the incidence of polyneuropathy in the myotonic dystrophies. Various studies in myasthenia gravis, including those concerning antibody testing, clinical features, and quality of life are also reviewed as are recent findings in congenital myasthenic syndromes. Finally, 2 studies concerning polyneuropathy are discussed, including one on the association of polyneuropathy in patients with the metabolic syndrome and one on laboratory testing in patients with otherwise idiopathic small fiber polyneuropathy.
Collapse
|
48
|
Aran A, Segel R, Kaneshige K, Gulsuner S, Renbaum P, Oliphant S, Meirson T, Weinberg-Shukron A, Hershkovitz Y, Zeligson S, Lee MK, Samson AO, Parsons SM, King MC, Levy-Lahad E, Walsh T. Vesicular acetylcholine transporter defect underlies devastating congenital myasthenia syndrome. Neurology 2017; 88:1021-1028. [PMID: 28188302 DOI: 10.1212/wnl.0000000000003720] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/21/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To identify the genetic basis of a recessive congenital neurologic syndrome characterized by severe hypotonia, arthrogryposis, and respiratory failure. METHODS Identification of the responsible gene by exome sequencing and assessment of the effect of the mutation on protein stability in transfected rat neuronal-like PC12A123.7 cells. RESULTS Two brothers from a nonconsanguineous Yemeni Jewish family manifested at birth with severe hypotonia and arthrogryposis. The older brother died of respiratory failure at 5 days of age. The proband, now 4.5 years old, has been mechanically ventilated since birth with virtually no milestones achievement. Whole exome sequencing revealed homozygosity of SLC18A3 c.1078G>C, p.Gly360Arg in the affected brothers but not in other family members. SLC18A3 p.Gly360Arg is not reported in world populations but is present at a carrier frequency of 1:30 in healthy Yemeni Jews. SLC18A3 encodes the vesicular acetylcholine transporter (VAChT), which loads newly synthesized acetylcholine from the neuronal cytoplasm into synaptic vesicles. Mice that are VAChT-null have been shown to die at birth of respiratory failure. In human VAChT, residue 360 is located in a conserved region and substitution of arginine for glycine is predicted to disrupt proper protein folding and membrane embedding. Stable transfection of wild-type and mutant human VAChT into neuronal-like PC12A123.7 cells revealed similar mRNA levels, but undetectable levels of the mutant protein, suggesting post-translational degradation of mutant VAChT. CONCLUSION Loss of function of VAChT underlies severe arthrogryposis and respiratory failure. While most congenital myasthenic syndromes are caused by defects in postsynaptic proteins, VAChT deficiency is a presynaptic myasthenic syndrome.
Collapse
Affiliation(s)
- Adi Aran
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Reeval Segel
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Kota Kaneshige
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Suleyman Gulsuner
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Paul Renbaum
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Scott Oliphant
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Tomer Meirson
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Ariella Weinberg-Shukron
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Yair Hershkovitz
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Sharon Zeligson
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Ming K Lee
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Abraham O Samson
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Stanley M Parsons
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Mary-Claire King
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| | - Ephrat Levy-Lahad
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle.
| | - Tom Walsh
- From the Neuropediatric Unit (A.A.) and Medical Genetics (R.S., P.R., A.W.-S., S.Z., E.L.-L.), Shaare Zedek Medical Center; Hebrew University-Hadassah School of Medicine (A.A., R.S., A.W.-S., E.L.-L.), Jerusalem, Israel; Department of Chemistry and Biochemistry (K.K., S.O., S.M.P.), University of California, Santa Barbara; Faculty of Medicine (T.M., Y.H., A.O.S.), Bar Ilan University, Safed, Israel; and Departments of Medicine and Genome Sciences (S.G., M.K.L., M.-C.K., T.W.), University of Washington, Seattle
| |
Collapse
|
49
|
Shen XM, Scola RH, Lorenzoni PJ, Kay CSK, Werneck LC, Brengman J, Selcen D, Engel AG. Novel synaptobrevin-1 mutation causes fatal congenital myasthenic syndrome. Ann Clin Transl Neurol 2017; 4:130-138. [PMID: 28168212 PMCID: PMC5288468 DOI: 10.1002/acn3.387] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 12/01/2022] Open
Abstract
Objective To identify the molecular basis and elucidate the pathogenesis of a fatal congenital myasthenic syndrome. Methods We performed clinical electrophysiology studies, exome and Sanger sequencing, and analyzed functional consequences of the identified mutation. Results Clinical electrophysiology studies of the patient revealed several‐fold potentiation of the evoked muscle action potential by high frequency nerve stimulation pointing to a presynaptic defect. Exome sequencing identified a homozygous c.340delA frameshift mutation in synaptobrevin 1 (SYB1), one of the three SNARE proteins essential for synaptic vesicle exocytosis. Analysis of both human spinal cord gray matter and normal human muscle revealed expression of the SYB1A and SYB1D isoforms, predicting expression of one or both isoforms in the motor nerve terminal. The identified mutation elongates the intravesicular C‐terminus of the A isoform from 5 to 71, and of the D isoform from 4 to 31 residues. Transfection of either mutant isoform into bovine chromaffin cells markedly reduces depolarization‐evoked exocytosis, and transfection of either mutant isoform into HEK cells significantly decreases expression of either mutant compared to wild type. Interpretation The mutation is pathogenic because elongation of the intravesicular C‐terminus of the A and D isoforms increases the energy required to move their C‐terminus into the synaptic vesicle membrane, a key step for fusion of the synaptic vesicle with the presynaptic membrane, and because it is predicted to reduce expression of either isoform in the nerve terminal.
Collapse
Affiliation(s)
- Xin-Ming Shen
- Department of Neurology and Muscle Research Laboratory Mayo Clinic Rochester Minnesota 55905
| | - Rosana H Scola
- Service of Neuromuscular Disorders Division of Neurology of Hospital de Clínicas (UFPR) Curitiba 80060-900 Brazil
| | - Paulo J Lorenzoni
- Service of Neuromuscular Disorders Division of Neurology of Hospital de Clínicas (UFPR) Curitiba 80060-900 Brazil
| | - Cláudia S K Kay
- Service of Neuromuscular Disorders Division of Neurology of Hospital de Clínicas (UFPR) Curitiba 80060-900 Brazil
| | - Lineu C Werneck
- Service of Neuromuscular Disorders Division of Neurology of Hospital de Clínicas (UFPR) Curitiba 80060-900 Brazil
| | - Joan Brengman
- Department of Neurology and Muscle Research Laboratory Mayo Clinic Rochester Minnesota 55905
| | - Duygu Selcen
- Department of Neurology and Muscle Research Laboratory Mayo Clinic Rochester Minnesota 55905
| | - Andrew G Engel
- Department of Neurology and Muscle Research Laboratory Mayo Clinic Rochester Minnesota 55905
| |
Collapse
|