Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 29(14): 2325-2336, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32543656

RESUMEN

Congenital myasthenic syndromes (CMS) are characterized by fatigable muscle weakness resulting from impaired neuromuscular transmission. ß2-adrenergic agonists are an effective treatment for DOK7-CMS. DOK7 is a component within the AGRN-LRP4-MUSK-DOK7 signalling pathway that is key for the formation and maintenance of the synaptic structure of the neuromuscular junction (NMJ). The precise mechanism of action of ß2-adrenergic agonists at the NMJ is not fully understood. In this study, we investigated whether ß2-adrenergic agonists improve both neurotransmission and structural integrity of the NMJ in a mouse model of DOK7-CMS. Ex-vivo electrophysiological techniques and microscopy of the NMJ were used to study the effect of salbutamol, a ß2-adrenergic agonist, on synaptic structure and function. DOK7-CMS model mice displayed a severe phenotype with reduced weight gain and perinatal lethality. Salbutamol treatment improved weight gain and survival in DOK7 myasthenic mice. Model animals had fewer active NMJs, detectable by endplate recordings, compared with age-matched wild-type littermates. Salbutamol treatment increased the number of detectable NMJs during endplate recording. Correspondingly, model mice had fewer acetylcholine receptor-stained NMJs detected by fluorescent labelling, but following salbutamol treatment an increased number were detectable. The data demonstrate that salbutamol can prolong survival and increase NMJ number in a severe model of DOK7-CMS.


Asunto(s)
Albuterol/farmacología , Proteínas Musculares/genética , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Unión Neuromuscular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/patología , Unión Neuromuscular/metabolismo , Embarazo , Receptores Colinérgicos/genética , Transducción de Señal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
2.
Proc Natl Acad Sci U S A ; 116(42): 21228-21235, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570625

RESUMEN

Charge selectivity forms the basis of cellular excitation or inhibition by Cys-loop ligand-gated ion channels (LGICs), and is essential for physiological receptor function. There are no reports of naturally occurring mutations in LGICs associated with the conversion of charge selectivity. Here, we report on a CHRNA1 mutation (α1Leu251Arg) in a patient with congenital myasthenic syndrome associated with transformation of the muscle acetylcholine receptor (AChR) into an inhibitory channel. Performing patch-clamp experiments, the AChR was found to be converted into chloride conductance at positive potentials, whereas whole-cell currents at negative potentials, although markedly reduced, were still carried by sodium. Umbrella sampling molecular dynamics simulations revealed constriction of the channel pore radius to 2.4 Å as a result of the mutation, which required partial desolvation of the ions in order to permeate the pore. Ion desolvation was associated with an energetic penalty that was compensated for by the favorable electrostatic interaction of the positively charged arginines with chloride. These findings reveal a mechanism for the transformation of the muscle AChR into an inhibitory channel in a clinical context.


Asunto(s)
Acetilcolina/metabolismo , Cloruros/metabolismo , Músculos/metabolismo , Mutación/genética , Receptores Colinérgicos/metabolismo , Línea Celular , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Síndromes Miasténicos Congénitos/metabolismo , Técnicas de Placa-Clamp/métodos , Receptores Nicotínicos/metabolismo , Sodio/metabolismo
3.
Hum Mutat ; 41(3): 619-631, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31765060

RESUMEN

MUSK encodes the muscle-specific receptor tyrosine kinase (MuSK), a key component of the agrin-LRP4-MuSK-DOK7 signaling pathway, which is essential for the formation and maintenance of highly specialized synapses between motor neurons and muscle fibers. We report a patient with severe early-onset congenital myasthenic syndrome and two novel missense mutations in MUSK (p.C317R and p.A617V). Functional studies show that MUSK p.C317R, located at the frizzled-like cysteine-rich domain of MuSK, disrupts an integral part of MuSK architecture resulting in ablated MuSK phosphorylation and acetylcholine receptor (AChR) cluster formation. MUSK p.A617V, located at the kinase domain of MuSK, enhances MuSK phosphorylation resulting in anomalous AChR cluster formation. The identification and evidence for pathogenicity of MUSK mutations supported the initiation of treatment with ß2-adrenergic agonists with a dramatic improvement of muscle strength in the patient. This work suggests uncharacterized mechanisms in which control of the precise level of MuSK phosphorylation is crucial in governing synaptic structure.


Asunto(s)
Mutación , Síndromes Miasténicos Congénitos/diagnóstico , Síndromes Miasténicos Congénitos/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Colinérgicos/genética , Sinapsis/genética , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Alelos , Sustitución de Aminoácidos , Animales , Sistemas CRISPR-Cas , Línea Celular , Análisis Mutacional de ADN , Femenino , Marcación de Gen , Humanos , Ratones , Modelos Moleculares , Conformación Molecular , Proteínas Musculares/metabolismo , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Síndromes Miasténicos Congénitos/metabolismo , Linaje , Fosforilación , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/química , Receptores Colinérgicos/metabolismo , Relación Estructura-Actividad , Sinapsis/metabolismo
4.
J Neurol Neurosurg Psychiatry ; 91(5): 526-532, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32165373

RESUMEN

OBJECTIVE: Direct inhibition of acetylcholine receptor (AChR) function by autoantibodies (Abs) is considered a rare pathogenic mechanism in myasthenia gravis (MG), but is usually studied on AChRs expressed in cell lines, rather than tightly clustered by the intracellular scaffolding protein, rapsyn, as at the intact neuromuscular junction. We hypothesised that clustered AChRs would provide a better target for investigating the functional effects of AChR-Abs. METHODS: Acetylcholine-induced currents were measured using whole-cell patch clamping and a fast perfusion system to assess fast (<2 min) functional effects of the serum samples. The sensitivity, specificity and rapidity of the system were first demonstrated by applying maternal AChR-Ab positive plasmas known to inhibit fetal AChR function in TE671 cells. Eleven previously untested AChR-Ab positive MG sera, 10 AChR-Ab negative MG sera and 5 healthy control sera were then applied to unclustered and rapsyn-clustered human adult AChRs in CN21 cells. RESULTS: The maternal AChR-Ab positive plasmas reduced fetal AChR currents, but not adult AChR currents, by >80% within 100 s. Only 2/11 AChR-Ab positive sera inhibited AChR currents in unclustered AChRs, but 6/11 AChR-Ab positive sera compared with none of the 10 AChR-Ab negative sera (p=0.0020) inhibited rapsyn-clustered AChR currents, and current inhibition by the AChR-Ab positive sera was greater when the AChRs were clustered (p=0.0385). None of the sera had detectable effects on desensitisation or recovery from desensitisation. CONCLUSION: These results show that antibodies can inhibit AChR function rapidly and demonstrate the importance of clustering in exploring pathogenic disease mechanisms of MG Abs.


Asunto(s)
Autoanticuerpos/inmunología , Proteínas Musculares/inmunología , Miastenia Gravis/inmunología , Receptores Colinérgicos/inmunología , Adolescente , Adulto , Anciano , Bungarotoxinas/farmacología , Línea Celular , Fenómenos Electrofisiológicos , Femenino , Fluoxetina/farmacología , Humanos , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Miastenia Gravis/etiología , Técnicas de Placa-Clamp , Receptores Colinérgicos/efectos de los fármacos , Adulto Joven
5.
Brain ; 142(12): 3713-3727, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31633155

RESUMEN

Acetylcholine receptor deficiency is the most common form of the congenital myasthenic syndromes, a heterogeneous collection of genetic disorders of neuromuscular transmission characterized by fatiguable muscle weakness. Most patients with acetylcholine receptor deficiency respond well to acetylcholinesterase inhibitors; however, in some cases the efficacy of acetylcholinesterase inhibitors diminishes over time. Patients with acetylcholine receptor deficiency can also benefit from the addition of a ß2-adrenergic receptor agonist to their medication. The working mechanism of ß2-adrenergic agonists in myasthenic patients is not fully understood. Here, we report the long-term follow-up for the addition of ß2-adrenergic agonists for a cohort of patients with acetylcholine receptor deficiency on anticholinesterase medication that demonstrates a sustained quantitative improvement. Coincidently we used a disease model to mirror the treatment of acetylcholine receptor deficiency, and demonstrate improved muscle fatigue, improved neuromuscular transmission and improved synaptic structure resulting from the addition of the ß2-adrenergic agonist salbutamol to the anticholinesterase medication pyridostigmine. Following an initial improvement in muscle fatiguability, a gradual decline in the effect of pyridostigmine was observed in mice treated with pyridostigmine alone (P < 0.001). Combination therapy with pyridostigmine and salbutamol counteracted this decline (P < 0.001). Studies of compound muscle action potential decrement at high nerve stimulation frequencies (P < 0.05) and miniature end-plate potential amplitude analysis (P < 0.01) showed an improvement in mice following combination therapy, compared to pyridostigmine monotherapy. Pyridostigmine alone reduced postsynaptic areas (P < 0.001) and postsynaptic folding (P < 0.01). Combination therapy increased postsynaptic area (P < 0.001) and promoted the formation of postsynaptic junctional folds (P < 0.001), in particular in fast-twitch muscles. In conclusion, we demonstrate for the first time how the improvement seen in patients from adding salbutamol to their medication can be explained in an experimental model of acetylcholine receptor deficiency, the most common form of congenital myasthenic syndrome. Salbutamol enhances neuromuscular junction synaptic structure by counteracting the detrimental effects of long-term acetylcholinesterase inhibitors on the postsynaptic neuromuscular junction. The results have implications for both autoimmune and genetic myasthenias where anticholinesterase medication is a standard treatment.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Albuterol/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Unión Neuromuscular/efectos de los fármacos , Bromuro de Piridostigmina/uso terapéutico , Potenciales de Acción/fisiología , Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Albuterol/uso terapéutico , Animales , Inhibidores de la Colinesterasa/farmacología , Modelos Animales de Enfermedad , Humanos , Ratones , Bromuro de Piridostigmina/farmacología , Transmisión Sináptica/efectos de los fármacos
6.
Brain ; 142(6): 1547-1560, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31081514

RESUMEN

Next generation sequencing techniques were recently used to show mutations in COL13A1 cause synaptic basal lamina-associated congenital myasthenic syndrome type 19. Animal studies showed COL13A1, a synaptic extracellular-matrix protein, is involved in the formation and maintenance of the neuromuscular synapse that appears independent of the Agrin-LRP4-MuSK-DOK7 acetylcholine receptor clustering pathway. Here, we report the phenotypic spectrum of 16 patients from 11 kinships harbouring homozygous or heteroallelic mutations in COL13A1. Clinical presentation was mostly at birth with hypotonia and breathing and feeding difficulties often requiring ventilation and artificial feeding. Respiratory crisis related to recurrent apnoeas, sometimes triggered by chest infections, were common early in life but resolved over time. The predominant pattern of muscle weakness included bilateral ptosis (non-fatigable in adulthood), myopathic facies and marked axial weakness, especially of neck flexion, while limb muscles were less involved. Other features included facial dysmorphism, skeletal abnormalities and mild learning difficulties. All patients tested had results consistent with abnormal neuromuscular transmission. Muscle biopsies were within normal limits or showed non-specific changes. Muscle MRI and serum creatine kinase levels were normal. In keeping with COL13A1 mutations affecting both synaptic structure and presynaptic function, treatment with 3,4-diaminopyridine and salbutamol resulted in motor and respiratory function improvement. In non-treated cases, disease severity and muscle strength improved gradually over time and several adults recovered normal muscle strength in the limbs. In summary, patients with COL13A1 mutations present mostly with severe early-onset myasthenic syndrome with feeding and breathing difficulties. Axial weakness is greater than limb weakness. Disease course improves gradually over time, which could be consistent with the less prominent role of COL13A1 once the neuromuscular junction is mature. This report emphasizes the role of collagens at the human muscle endplate and should facilitate the recognition of this disorder, which can benefit from pharmacological treatment.


Asunto(s)
Colágeno Tipo XIII/genética , Proteínas Musculares/genética , Síndromes Miasténicos Congénitos/genética , Unión Neuromuscular/metabolismo , Transmisión Sináptica/genética , Adolescente , Adulto , Niño , Femenino , Homocigoto , Humanos , Masculino , Músculo Esquelético/patología , Mutación/genética , Síndromes Miasténicos Congénitos/diagnóstico , Unión Neuromuscular/genética , Sinapsis/genética , Adulto Joven
7.
J Physiol ; 597(14): 3713-3725, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31158924

RESUMEN

KEY POINTS: The physiological significance of the developmental switch from fetal to adult acetylcholine receptors in muscle (AChRs) and the functional impact of AChR clustering by rapsyn are not well studied. Using patch clamp experiments, we show that recovery from desensitization is faster in the adult AChR isoform. Recovery from desensitization is determined by the AChR isoform-specific cytoplasmic M3-M4 domain. The co-expression of rapsyn in muscle cells induced AChR clustering and facilitated recovery from desensitization in both fetal and adult AChRs. In fetal AChRs, facilitation of recovery kinetics by rapsyn was independent of AChR clustering. These effects could be crucial adaptations to motor neuron firing rates, which, in rodents, have been shown to increase around the time of birth when AChRs cluster at the developing neuromuscular junctions. ABSTRACT: The neuromuscular junction (NMJ) is the site of a number of autoimmune and genetic disorders, many involving the muscle-type nicotinic acetylcholine receptor (AChR), although there are aspects of normal NMJ development and function that need to be better understood. In particular, there are still questions regarding the implications of the developmental switch from fetal to adult AChRs, as well as how their functions might be modified by rapsyn that clusters the AChRs. Desensitization of human muscle AChRs was investigated using the patch clamp technique to measure whole-cell currents in muscle-type (TE671/CN21) and non-muscle (HEK293) cell lines expressing either fetal or adult AChRs. Desensitization time constants were similar with both AChR isoforms but recovery time constants were shorter in cells expressing adult compared to fetal AChRs (P < 0.0001). Chimeric experiments showed that recovery from desensitization was determined by the M3-M4 cytoplasmic loops of the γ- and ε-subunits. Expression of rapsyn in TE671/CN21 cells induced AChR aggregation and also, surprisingly, shortened recovery time constants in both fetal and adult AChRs. However, this was not dependent on clustering because rapsyn also facilitated recovery from desensitization in HEK293 cells expressing a δ-R375H AChR mutant that did not form clusters in C2C12 myotubes. Thus, rapsyn interactions with AChRs lead not only to clustering, but also to a clustering independent faster recovery from desensitization. Both effects of rapsyn could be a necessary adjustment to the motor neuron firing rates that increase around the time of birth.


Asunto(s)
Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/farmacología , Receptores Nicotínicos/metabolismo , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/metabolismo , Receptores Colinérgicos/metabolismo
8.
Am J Hum Genet ; 97(6): 878-85, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26626625

RESUMEN

The neuromuscular junction (NMJ) consists of a tripartite synapse with a presynaptic nerve terminal, Schwann cells that ensheathe the terminal bouton, and a highly specialized postsynaptic membrane. Synaptic structural integrity is crucial for efficient signal transmission. Congenital myasthenic syndromes (CMSs) are a heterogeneous group of inherited disorders that result from impaired neuromuscular transmission, caused by mutations in genes encoding proteins that are involved in synaptic transmission and in forming and maintaining the structural integrity of NMJs. To identify further causes of CMSs, we performed whole-exome sequencing (WES) in families without an identified mutation in known CMS-associated genes. In two families affected by a previously undefined CMS, we identified homozygous loss-of-function mutations in COL13A1, which encodes the alpha chain of an atypical non-fibrillar collagen with a single transmembrane domain. COL13A1 localized to the human muscle motor endplate. Using CRISPR-Cas9 genome editing, modeling of the COL13A1 c.1171delG (p.Leu392Sfs(∗)71) frameshift mutation in the C2C12 cell line reduced acetylcholine receptor (AChR) clustering during myotube differentiation. This highlights the crucial role of collagen XIII in the formation and maintenance of the NMJ. Our results therefore delineate a myasthenic disorder that is caused by loss-of-function mutations in COL13A1, encoding a protein involved in organization of the NMJ, and emphasize the importance of appropriate symptomatic treatment for these individuals.


Asunto(s)
Colágeno Tipo XIII/genética , Mutación , Síndromes Miasténicos Congénitos/genética , Mioblastos/metabolismo , Unión Neuromuscular/metabolismo , Adulto , Animales , Línea Celular , Preescolar , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Colágeno Tipo XIII/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Exoma , Femenino , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Masculino , Ratones , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/patología , Mioblastos/patología , Unión Neuromuscular/crecimiento & desarrollo , Unión Neuromuscular/patología , Linaje , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/patología , Transmisión Sináptica
9.
Am J Pathol ; 186(10): 2559-68, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27658713

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease mediated by autoantibodies that target proteins at the neuromuscular junction, primarily the acetylcholine receptor (AChR) and the muscle-specific kinase. Because downstream of kinase 7 (Dok-7) is essential for the full activation of muscle-specific kinase and consequently for dense clustering of AChRs, we hypothesized that reduced levels of Dok-7 increase the susceptibility to passive transfer MG. To test this hypothesis, Dok-7 expression was reduced by transfecting shRNA-coding plasmids into the tibialis anterior muscle of adult rats by in vivo electroporation. Subclinical MG was subsequently induced with a low dose of anti-AChR monoclonal antibody 35. Neuromuscular transmission was significantly impaired in Dok-7-siRNA-electroporated legs compared with the contralateral control legs, which correlated with a reduction of AChR protein levels at the neuromuscular junction (approximately 25%) in Dok-7-siRNA-electroporated muscles, compared with contralateral control muscles. These results suggest that a reduced expression of Dok-7 may play a role in the susceptibility to passive transfer MG, by rendering AChR clusters less resistant to the autoantibody attack.


Asunto(s)
Autoanticuerpos/inmunología , Proteínas Musculares/genética , Miastenia Gravis Autoinmune Experimental/genética , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Femenino , Silenciador del Gen , Genes Reporteros , Células HEK293 , Humanos , Proteínas Musculares/metabolismo , Músculo Esquelético/inmunología , Músculo Esquelético/fisiopatología , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/fisiopatología , Unión Neuromuscular/inmunología , Unión Neuromuscular/fisiopatología , Ratas , Ratas Endogámicas Lew , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Transmisión Sináptica
10.
BMC Neurol ; 16(1): 195, 2016 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-27717316

RESUMEN

BACKGROUND: Congenital myasthenic syndromes (CMSs) occur as a result of genetic mutations that cause aberrations in structure and/or function of proteins involved in neuromuscular transmission. Acetylcholine receptor epsilon (ε) subunit (CHRNE) gene mutations account for about 30-50 % of genetically diagnosed cases. We report a rare CHRNE gene mutation in a South Asian female with CMS. CASE PRESENTATION: A 17-year-old Maldivian female presented with bilateral partial ptosis, fatigable proximal muscle weakness and slurring of speech noted since the age of 2 years. She could not run, had difficulty negotiating stairs and rising from a seated position, and fatigues when speaking at length. Her birth and past medical histories were otherwise unremarkable. There is no parental consanguinity or family history of muscle disorders. On examination, she had a BMI of 18 kg/m2, bilateral fatigable partial ptosis, complete external ophthalmoplegia and fatigable proximal muscle weakness (MRC grade 4/5). Apart from spinal scoliosis the rest of the examination was normal. Haematological and biochemical investigations including serum lactate level and thyroid functions were normal. Acetylcholine receptor antibodies and muscle specific kinase antibodies were not detected in serum. Repetitive nerve stimulation showed marked decrement (>30 %) in nerve-muscle pairs in the face and forearm. Her DNA sequencing revealed a c.183-187dupCTCAC mutation in CHRNE. She remained functionally independent on pyridostigmine treatment. CONCLUSIONS: This case describes a rare mutation of the CHRNE gene in CMS and highlights the relevance of genetic diagnosis in CMS. It further adds to map the occurrence of such mutations in Asian populations.


Asunto(s)
Mutación , Síndromes Miasténicos Congénitos/genética , Receptores Nicotínicos/genética , Adolescente , Secuencia de Bases , Femenino , Humanos
11.
Hum Mol Genet ; 22(14): 2905-13, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23569079

RESUMEN

Mutations in GFPT1 underlie a congenital myasthenic syndrome (CMS) characterized by a limb-girdle pattern of muscle weakness. Glutamine-fructose-6-phosphate transaminase 1 (GFPT1) is a key rate-limiting enzyme in the hexosamine biosynthetic pathway providing building blocks for the glycosylation of proteins and lipids. It is expressed ubiquitously and it is not readily apparent why mutations in this gene should cause a syndrome with symptoms restricted to muscle and, in particular, to the neuromuscular junction. Data from a muscle biopsy obtained from a patient with GFPT1 mutations indicated that there were reduced endplate acetylcholine receptors. We, therefore, further investigated the relationship between identified mutations in GFPT1 and expression of the muscle acetylcholine receptor. Cultured myotubes derived from two patients with GFPT1 mutations showed a significant reduction in cell-surface AChR expression (Pt1 P < 0.0001; Pt2 P = 0.0097). Inhibition of GFPT1 enzymatic activity or siRNA silencing of GFPT1 expression both resulted in reduced AChR cell-surface expression. Western blot and gene-silencing experiments indicate this is due to reduced steady-state levels of AChR α, δ, ε, but not ß subunits rather than altered transcription of AChR-subunit RNA. Uridine diphospho-N-acetylglucosamine, a product of the hexosamine synthetic pathway, acts as a substrate at an early stage in the N-linked glycosylation pathway. Similarity between CMS due to GFPT1 mutations and CMS due to DPAGT1 mutations would suggest that reduced endplate AChR due to defective N-linked glycosylation is a primary disease mechanism in this disorder.


Asunto(s)
Regulación hacia Abajo , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética , Mutación , Síndromes Miasténicos Congénitos/enzimología , Receptores Colinérgicos/genética , Células Cultivadas , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/metabolismo , Glicosilación , Humanos , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/metabolismo , Receptores Colinérgicos/metabolismo
12.
Am J Hum Genet ; 91(1): 193-201, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22742743

RESUMEN

Congenital myasthenic syndromes are a heterogeneous group of inherited disorders that arise from impaired signal transmission at the neuromuscular synapse. They are characterized by fatigable muscle weakness. We performed whole-exome sequencing to determine the underlying defect in a group of individuals with an inherited limb-girdle pattern of myasthenic weakness. We identify DPAGT1 as a gene in which mutations cause a congenital myasthenic syndrome. We describe seven different mutations found in five individuals with DPAGT1 mutations. The affected individuals share a number of common clinical features, including involvement of proximal limb muscles, response to treatment with cholinesterase inhibitors and 3,4-diaminopyridine, and the presence of tubular aggregates in muscle biopsies. Analyses of motor endplates from two of the individuals demonstrate a severe reduction of endplate acetylcholine receptors. DPAGT1 is an essential enzyme catalyzing the first committed step of N-linked protein glycosylation. Our findings underscore the importance of N-linked protein glycosylation for proper functioning of the neuromuscular junction. Using the DPAGT1-specific inhibitor tunicamycin, we show that DPAGT1 is required for efficient glycosylation of acetylcholine-receptor subunits and for efficient export of acetylcholine receptors to the cell surface. We suggest that the primary pathogenic mechanism of DPAGT1 mutations is reduced levels of acetylcholine receptors at the endplate region. These individuals share clinical features similar to those of congenital myasthenic syndrome due to GFPT1 mutations, and their disorder might be part of a larger subgroup comprising the congenital myasthenic syndromes that result from defects in the N-linked glycosylation pathway and that manifest through impaired neuromuscular transmission.


Asunto(s)
Síndromes Miasténicos Congénitos/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , 4-Aminopiridina/análogos & derivados , 4-Aminopiridina/farmacología , Adulto , Amifampridina , Inhibidores de la Colinesterasa/uso terapéutico , Femenino , Glicosilación , Humanos , Extremidad Inferior , Masculino , Persona de Mediana Edad , Placa Motora/metabolismo , Mutación , Síndromes Miasténicos Congénitos/patología , Unión Neuromuscular/metabolismo , Receptores Colinérgicos/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/antagonistas & inhibidores , Tunicamicina/farmacología
13.
Hum Mol Genet ; 21(17): 3765-75, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661499

RESUMEN

Congenital myasthenic syndromes (CMS) are a group of inherited diseases that affect synaptic transmission at the neuromuscular junction and result in fatiguable muscle weakness. A subgroup of CMS patients have a recessively inherited limb-girdle pattern of weakness caused by mutations in DOK7. DOK7 encodes DOK7, an adaptor protein that is expressed in the skeletal muscle and heart and that is essential for the development and maintenance of the neuromuscular junction. We have screened the DOK7 gene for mutations by polymerase chain reaction amplification and bi-directional sequencing of exonic and promoter regions and performed acetylcholine receptor (AChR) clustering assays and used exon trapping to determine the pathogenicity of detected variants. Approximately 18% of genetically diagnosed CMSs in the UK have mutations in DOK7, with mutations in this gene identified in more than 60 kinships to date. Thirty-four different pathogenic mutations were identified as well as 27 variants likely to be non-pathogenic. An exon 7 frameshift duplication c.1124_1127dupTGCC is commonly found in at least one allele. We analyse the effect of the common frameshift c.1124_1127dupTGCC and show that 10/11 suspected missense mutations have a deleterious effect on AChR clustering. We identify for the first time homozygous or compound heterozygous mutations that are localized 5' to exon 7. In addition, three silent variants in the N-terminal half of DOK7 are predicted to alter the splicing of the DOK7 RNA transcript. The DOK7 gene is highly polymorphic, and within these many variants, we define a spectrum of mutations that can underlie DOK7 CMS that will inform in managing this disorder.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas Musculares/genética , Mutación/genética , Síndromes Miasténicos Congénitos/genética , Unión Neuromuscular/genética , Sinapsis/genética , Sinapsis/patología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Exones/genética , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Musculares/química , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación Missense/genética , Síndromes Miasténicos Congénitos/patología , Sistemas de Lectura Abierta/genética , Receptores Colinérgicos/metabolismo , Alineación de Secuencia , Transfección
14.
J Anat ; 224(1): 29-35, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23458718

RESUMEN

MuSK myasthenia gravis is a rare, severe autoimmune disease of the neuromuscular junction, only identified in 2001, with unclear pathogenic mechanisms. In this review we describe the clinical aspects that distinguish MuSK MG from AChR MG, review what is known about the role of MuSK in the development and function of the neuromuscular junction, and discuss the data that address how the antibodies to MuSK lead to neuromuscular transmission failure.


Asunto(s)
Miastenia Gravis/fisiopatología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Colinérgicos/fisiología , Autoanticuerpos/fisiología , Humanos , Miastenia Gravis/inmunología , Unión Neuromuscular/inmunología , Unión Neuromuscular/fisiología , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología
15.
Brain ; 136(Pt 3): 944-56, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23404334

RESUMEN

Congenital myasthenic syndromes are a heterogeneous group of inherited disorders that arise from impaired signal transmission at the neuromuscular synapse. They are characterized by fatigable muscle weakness. We performed linkage analysis, whole-exome and whole-genome sequencing to determine the underlying defect in patients with an inherited limb-girdle pattern of myasthenic weakness. We identify ALG14 and ALG2 as novel genes in which mutations cause a congenital myasthenic syndrome. Through analogy with yeast, ALG14 is thought to form a multiglycosyltransferase complex with ALG13 and DPAGT1 that catalyses the first two committed steps of asparagine-linked protein glycosylation. We show that ALG14 is concentrated at the muscle motor endplates and small interfering RNA silencing of ALG14 results in reduced cell-surface expression of muscle acetylcholine receptor expressed in human embryonic kidney 293 cells. ALG2 is an alpha-1,3-mannosyltransferase that also catalyses early steps in the asparagine-linked glycosylation pathway. Mutations were identified in two kinships, with mutation ALG2p.Val68Gly found to severely reduce ALG2 expression both in patient muscle, and in cell cultures. Identification of DPAGT1, ALG14 and ALG2 mutations as a cause of congenital myasthenic syndrome underscores the importance of asparagine-linked protein glycosylation for proper functioning of the neuromuscular junction. These syndromes form part of the wider spectrum of congenital disorders of glycosylation caused by impaired asparagine-linked glycosylation. It is likely that further genes encoding components of this pathway will be associated with congenital myasthenic syndromes or impaired neuromuscular transmission as part of a more severe multisystem disorder. Our findings suggest that treatment with cholinesterase inhibitors may improve muscle function in many of the congenital disorders of glycosylation.


Asunto(s)
Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/fisiopatología , N-Acetilglucosaminiltransferasas/genética , Adolescente , Edad de Inicio , Secuencia de Bases , Western Blotting , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Síndromes Miasténicos Congénitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Adulto Joven
16.
bioRxiv ; 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38405691

RESUMEN

Congenital myasthenic syndromes (CMS) are a group of inherited disorders characterised by defective neuromuscular transmission and fatigable muscle weakness. Mutations in DOK7 , a gene encoding a post-synaptic protein crucial in the formation and stabilisation of the neuromuscular junction (NMJ), rank among the leading three prevalent causes of CMS in diverse populations globally. The majority of DOK7 CMS patients experience varying degrees of disability despite receiving optimised treatment, necessitating the development of improved therapeutic approaches. Here we executed a dose escalation pre-clinical trial using a DOK7-CMS mouse model to assess the efficacy of Amp-101, an innovative AAV gene replacement therapy. Amp-101 is based on AAVrh74 and contains human DOK7 cDNA under the control of a muscle-restricted promoter. We show that at doses 6x10 13 vg/kg and 1x10 14 vg/kg, Amp-101 generated enlarged NMJs and rescued the very severe phenotype of the model. Treated mice became at least as strong as WT littermates and the diaphragm and tibialis anterior muscles displayed robust expression of DOK7. This data suggests that Amp-101 is a promising candidate to move forward to clinic trials.

17.
J Neurol Neurosurg Psychiatry ; 84(10): 1119-25, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23447650

RESUMEN

BACKGROUND: A newly defined congenital myasthenic syndrome (CMS) caused by DPAGT1 mutations has recently been reported. While many other CMS-associated proteins have discrete roles localised to the neuromuscular junction, DPAGT1 is ubiquitously expressed, modifying many proteins, and as such is an unexpected cause of isolated neuromuscular involvement. METHODS: We present detailed clinical characteristics of five patients with CMS caused by DPAGT1 mutations. RESULTS: Patients have prominent limb girdle weakness and minimal craniobulbar symptoms. Tubular aggregates on muscle biopsy are characteristic but may not be apparent on early biopsies. Typical myasthenic features such as pyridostigmine and 3, 4- diaminopyridine responsiveness, and decrement on repetitive nerve stimulation are present. CONCLUSIONS: These patients mimic myopathic disorders and are likely to be under-diagnosed. The descriptions here should facilitate recognition of this disorder. In particular minimal craniobulbar involvement and tubular aggregates on muscle biopsy help to distinguish DPAGT1 CMS from the majority of other forms of CMS. Patients with DPAGT1 CMS share similar clinical features with patients who have CMS caused by mutations in GFPT1, another recently identified CMS subtype.


Asunto(s)
Análisis Mutacional de ADN , Síndromes Miasténicos Congénitos/genética , N-Acetilglucosaminiltransferasas/genética , 4-Aminopiridina/análogos & derivados , 4-Aminopiridina/uso terapéutico , Agonistas de Receptores Adrenérgicos beta 2 , Adulto , Edad de Inicio , Albuterol/uso terapéutico , Amifampridina , Biopsia , Inhibidores de la Colinesterasa/uso terapéutico , Diagnóstico Diferencial , Exoma , Femenino , Pruebas Genéticas , Glicosilación , Humanos , Masculino , Persona de Mediana Edad , Neuronas Motoras/fisiología , Músculo Esquelético/inervación , Músculo Esquelético/patología , Síndromes Miasténicos Congénitos/diagnóstico , Síndromes Miasténicos Congénitos/patología , Síndromes Miasténicos Congénitos/fisiopatología , Examen Neurológico , Unión Neuromuscular/fisiología , Fenotipo , Bloqueadores de los Canales de Potasio/uso terapéutico , Bromuro de Piridostigmina/uso terapéutico
18.
J Neuroimmunol ; 383: 578195, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37660538

RESUMEN

INTRODUCTION: Muscle specific kinase (MuSK) antibody positive myasthenia gravis (MG) often presents with a severe disease course and resistance to treatment. Treatment-refractory patients may respond to B cell depleting treatment methods. Our aim was to investigate whether inhibition of Fc receptor-like B (FCRLB) could effectively suppress autoimmunity without diminishing B cell counts in animal model of MG, a classical antibody-mediated autoimmune disease. METHODS: Experimental autoimmune MG was induced in Balb/C mice with two s.c. immunizations with recombinant human MuSK in complete Freund's adjuvant. FCRLB was silenced with a lentiviral particle transported shRNA in myasthenic mice with a single i.p. injection during second MuSK-immunization. Control immunized mice received scrambled shRNA or saline. Mice were observed for clinical parameters for 28 days and at termination, anti-MuSK IgG, neuromuscular junction (NMJ) deposits, muscle AChR expression and lymph node B and T cell ratios were assessed by ELISA, immunofluorescence, immunoblotting and flow cytometry, respectively. RESULTS: FCRLB shRNA-treated mice showed no muscle weakness or weight loss at termination. Also, they exhibited higher grip strength and muscle AChR levels, lower anti-MuSK IgG and NMJ IgG/C3 levels than control mice. Flow cytometry analysis showed that ratios of major effector lymph node B and T cell populations were not altered by FCRLB silencing. However, regulatory T and CD19 + CD5+ B cell ratios were decreased in FCRLB shRNA-group. CONCLUSION: Our results provide evidence regarding involvement and therapeutic value of FCRLB in MuSK-MG. Silencing of FCRLB appears to substantially inhibit antibody production without interfering with survival of major lymphocyte populations.


Asunto(s)
Inmunoglobulina G , Miastenia Gravis Autoinmune Experimental , Humanos , Ratones , Animales , Proteínas Tirosina Quinasas Receptoras , Unión Neuromuscular , Inmunización/métodos , Autoanticuerpos
19.
Artículo en Inglés | MEDLINE | ID: mdl-37582613

RESUMEN

BACKGROUND AND OBJECTIVES: Up to 50% of patients with myasthenia gravis (MG) without acetylcholine receptor antibodies (AChR-Abs) have antibodies to muscle-specific kinase (MuSK). Most MuSK antibodies (MuSK-Abs) are IgG4 and inhibit agrin-induced MuSK phosphorylation, leading to impaired clustering of AChRs at the developing or mature neuromuscular junction. However, IgG1-3 MuSK-Abs also exist in MuSK-MG patients, and their potential mechanisms have not been explored fully. METHODS: C2C12 myotubes were exposed to MuSK-MG plasma IgG1-3 or IgG4, with or without purified agrin. MuSK, Downstream of Kinase 7 (DOK7), and ßAChR were immunoprecipitated and their phosphorylation levels identified by immunoblotting. Agrin and agrin-independent AChR clusters were measured by immunofluorescence and AChR numbers by binding of 125I-α-bungarotoxin. Transcriptomic analysis was performed on treated myotubes. RESULTS: IgG1-3 MuSK-Abs impaired AChR clustering without inhibiting agrin-induced MuSK phosphorylation. Moreover, the well-established pathway initiated by MuSK through DOK7, resulting in ßAChR phosphorylation, was not impaired by MuSK-IgG1-3 and was agrin-independent. Nevertheless, the AChR clusters did not form, and both the number of AChR microclusters that precede full cluster formation and the myotube surface AChRs were reduced. Transcriptomic analysis did not throw light on the pathways involved. However, the SHP2 inhibitor, NSC-87877, increased the number of microclusters and led to fully formed AChR clusters. DISCUSSION: MuSK-IgG1-3 is pathogenic but seems to act through a noncanonical pathway. Further studies should throw light on the mechanisms involved at the neuromuscular junction.


Asunto(s)
Miastenia Gravis , Proteínas Tirosina Quinasas Receptoras , Humanos , Agrina/farmacología , Inmunoglobulina G , Proteínas Musculares/metabolismo , Miastenia Gravis/tratamiento farmacológico , Fosforilación , Receptores Colinérgicos
20.
J Neuromuscul Dis ; 8(1): 163-167, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33216040

RESUMEN

Congenital myasthenic syndromes (CMS) result from genetic mutations that cause aberrations in structure and/or function of proteins involved in neuromuscular transmission. The slow-channel CMS (SCCMS) is an autosomal dominant postsynaptic defect caused by mutations in genes encoding alpha, beta, delta, or epsilon subunits of the acetylcholine receptor resulting in a functional defect which is an increase of the opening time of the receptor. We report a case of SCCMS due to a heterozygous mutation in the M2 domain of the AChR alpha subunit - CHRNA1:ENST00000348749.6:exon7:c.806T>G:p.Val269Gly and corresponding kinetic defect. A substitution of valine with phenylalanine in the same position has been previously described. This is the first reported case of a new CHRNA1 variant in a patient with SCCMS from South Asia. We also highlight the phenotype that would favour a genetic basis over an autoimmune one, in an adult presenting with fatigable weakness.


Asunto(s)
Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/fisiopatología , Receptores Nicotínicos/genética , Adulto , Humanos , Síndromes Miasténicos Congénitos/diagnóstico , Sri Lanka
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA