Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 60(10): 1026-1034, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37197783

RESUMEN

BACKGROUND: RNA polymerase III-related or 4H leukodystrophy (POLR3-HLD) is an autosomal recessive hypomyelinating leukodystrophy characterized by neurological dysfunction, hypodontia and hypogonadotropic hypogonadism. The disease is caused by biallelic pathogenic variants in POLR3A, POLR3B, POLR1C or POLR3K. Craniofacial abnormalities reminiscent of Treacher Collins syndrome have been originally described in patients with POLR3-HLD caused by biallelic pathogenic variants in POLR1C. To date, no published studies have appraised in detail the craniofacial features of patients with POLR3-HLD. In this work, the specific craniofacial characteristics of patients with POLR3-HLD associated with biallelic pathogenic variants in POLR3A, POLR3B and POLR1C are described. METHODS: The craniofacial features of 31 patients with POLR3-HLD were evaluated, and potential genotype-phenotype associations were evaluated. RESULTS: Various craniofacial abnormalities were recognized in this patient cohort, with each individual presenting at least one craniofacial abnormality. The most frequently identified features included a flat midface (61.3%), a smooth philtrum (58.0%) and a pointed chin (51.6%). In patients with POLR3B biallelic variants, a thin upper lip was frequent. Craniofacial anomalies involving the forehead were most commonly associated with biallelic variants in POLR3A and POLR3B while a higher proportion of patients with POLR1C biallelic variants demonstrated bitemporal narrowing. CONCLUSION: Through this study, we demonstrated that craniofacial abnormalities are common in patients with POLR3-HLD. This report describes in detail the dysmorphic features of POLR3-HLD associated with biallelic variants in POLR3A, POLR3B and POLR1C.


Asunto(s)
Enfermedades Desmielinizantes , Enfermedades Neurodegenerativas , Humanos , ARN Polimerasa III/genética , Patrón de Herencia , ARN Polimerasas Dirigidas por ADN/genética
2.
Am J Hum Genet ; 105(2): 384-394, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31256876

RESUMEN

Proteins anchored to the cell surface via glycosylphosphatidylinositol (GPI) play various key roles in the human body, particularly in development and neurogenesis. As such, many developmental disorders are caused by mutations in genes involved in the GPI biosynthesis and remodeling pathway. We describe ten unrelated families with bi-allelic mutations in PIGB, a gene that encodes phosphatidylinositol glycan class B, which transfers the third mannose to the GPI. Ten different PIGB variants were found in these individuals. Flow cytometric analysis of blood cells and fibroblasts from the affected individuals showed decreased cell surface presence of GPI-anchored proteins. Most of the affected individuals have global developmental and/or intellectual delay, all had seizures, two had polymicrogyria, and four had a peripheral neuropathy. Eight children passed away before four years old. Two of them had a clinical diagnosis of DOORS syndrome (deafness, onychodystrophy, osteodystrophy, mental retardation, and seizures), a condition that includes sensorineural deafness, shortened terminal phalanges with small finger and toenails, intellectual disability, and seizures; this condition overlaps with the severe phenotypes associated with inherited GPI deficiency. Most individuals tested showed elevated alkaline phosphatase, which is a characteristic of the inherited GPI deficiency but not DOORS syndrome. It is notable that two severely affected individuals showed 2-oxoglutaric aciduria, which can be seen in DOORS syndrome, suggesting that severe cases of inherited GPI deficiency and DOORS syndrome might share some molecular pathway disruptions.


Asunto(s)
Anomalías Craneofaciales/etiología , Glicosilfosfatidilinositoles/biosíntesis , Glicosilfosfatidilinositoles/deficiencia , Deformidades Congénitas de la Mano/etiología , Pérdida Auditiva Sensorineural/etiología , Discapacidad Intelectual/etiología , Manosiltransferasas/genética , Enfermedades Metabólicas/etiología , Mutación , Uñas Malformadas/etiología , Enfermedades del Sistema Nervioso Periférico/etiología , Convulsiones/patología , Adulto , Niño , Preescolar , Anomalías Craneofaciales/patología , Femenino , Glicosilfosfatidilinositoles/genética , Deformidades Congénitas de la Mano/patología , Pérdida Auditiva Sensorineural/patología , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/patología , Masculino , Enfermedades Metabólicas/patología , Uñas Malformadas/patología , Linaje , Enfermedades del Sistema Nervioso Periférico/patología , Convulsiones/genética , Índice de Severidad de la Enfermedad , Adulto Joven
3.
Mol Psychiatry ; 26(12): 7225-7246, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34127816

RESUMEN

CACNA1A deletions cause epilepsy, ataxia, and a range of neurocognitive deficits, including inattention, impulsivity, intellectual deficiency and autism. To investigate the underlying mechanisms, we generated mice carrying a targeted Cacna1a deletion restricted to parvalbumin-expressing (PV) neurons (PVCre;Cacna1ac/+) or to cortical pyramidal cells (PC) (Emx1Cre;Cacna1ac/+). GABA release from PV-expressing GABAergic interneurons (PV-INs) is reduced in PVCre;Cacna1ac/+ mutants, resulting in impulsivity, cognitive rigidity and inattention. By contrast, the deletion of Cacna1a in PCs does not impact cortical excitability or behaviour in Emx1Cre;Cacna1ac/+ mutants. A targeted Cacna1a deletion in the orbitofrontal cortex (OFC) results in reversal learning deficits while a medial prefrontal cortex (mPFC) deletion impairs selective attention. These deficits can be rescued by the selective chemogenetic activation of cortical PV-INs in the OFC or mPFC of PVCre;Cacna1ac/+ mutants. Thus, Cacna1a haploinsufficiency disrupts perisomatic inhibition in frontal cortical circuits, leading to a range of potentially reversible neurocognitive deficits.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Interneuronas , Trastornos del Neurodesarrollo , Parvalbúminas , Animales , Interneuronas/metabolismo , Ratones , Neuronas/metabolismo , Parvalbúminas/metabolismo , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo
4.
Hum Mol Genet ; 28(4): 584-597, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30335140

RESUMEN

Mutations in the Tre2/Bub2/Cdc16 (TBC)1 domain family member 24 (TBC1D24) gene are associated with a range of inherited neurological disorders, from drug-refractory lethal epileptic encephalopathy and DOORS syndrome (deafness, onychodystrophy, osteodystrophy, mental retardation, seizures) to non-syndromic hearing loss. TBC1D24 has been implicated in neuronal transmission and maturation, although the molecular function of the gene and the cause of the apparently complex disease spectrum remain unclear. Importantly, heterozygous TBC1D24 mutation carriers have also been reported with seizures, suggesting that haploinsufficiency for TBC1D24 is significant clinically. Here we have systematically investigated an allelic series of disease-associated mutations in neurons alongside a new mouse model to investigate the consequences of TBC1D24 haploinsufficiency to mammalian neurodevelopment and synaptic physiology. The cellular studies reveal that disease-causing mutations that disrupt either of the conserved protein domains in TBC1D24 are implicated in neuronal development and survival and are likely acting as loss-of-function alleles. We then further investigated TBC1D24 haploinsufficiency in vivo and demonstrate that TBC1D24 is also crucial for normal presynaptic function: genetic disruption of Tbc1d24 expression in the mouse leads to an impairment of endocytosis and an enlarged endosomal compartment in neurons with a decrease in spontaneous neurotransmission. These data reveal the essential role for TBC1D24 at the mammalian synapse and help to define common synaptic mechanisms that could underlie the varied effects of TBC1D24 mutations in neurological disease.


Asunto(s)
Proteínas Portadoras/genética , Anomalías Craneofaciales/genética , Epilepsia/genética , Deformidades Congénitas de la Mano/genética , Pérdida Auditiva Sensorineural/genética , Discapacidad Intelectual/genética , Uñas Malformadas/genética , Convulsiones/genética , Secuencia de Aminoácidos/genética , Animales , Anomalías Craneofaciales/fisiopatología , Modelos Animales de Enfermedad , Endocitosis/genética , Epilepsia/fisiopatología , Exoma/genética , Proteínas Activadoras de GTPasa , Regulación de la Expresión Génica , Deformidades Congénitas de la Mano/fisiopatología , Haploinsuficiencia , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Discapacidad Intelectual/fisiopatología , Proteínas de la Membrana , Ratones , Mutación , Uñas Malformadas/fisiopatología , Proteínas del Tejido Nervioso , Plasticidad Neuronal/genética , Neuronas/metabolismo , Neuronas/patología , Linaje , Convulsiones/fisiopatología
5.
Epilepsia ; 62(1): e13-e21, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33280099

RESUMEN

Chromosome 1q41-q42 deletion syndrome is a rare cause of intellectual disability, seizures, dysmorphology, and multiple anomalies. Two genes in the 1q41-q42 microdeletion, WDR26 and FBXO28, have been implicated in monogenic disease. Patients with WDR26 encephalopathy overlap clinically with those with 1q41-q42 deletion syndrome, whereas only one patient with FBXO28 encephalopathy has been described. Seizures are a prominent feature of 1q41-q42 deletion syndrome; therefore, we hypothesized that pathogenic FBXO28 variants cause developmental and epileptic encephalopathies (DEEs). We describe nine new patients with FBXO28 pathogenic variants (four missense, including one recurrent, three nonsense, and one frameshift) and analyze all 10 known cases to delineate the phenotypic spectrum. All patients had epilepsy and 9 of 10 had DEE, including infantile spasms (3) and a progressive myoclonic epilepsy (1). Median age at seizure onset was 22.5 months (range 8 months to 5 years). Nine of 10 patients had intellectual disability, which was profound in six of nine and severe in three of nine. Movement disorders occurred in eight of 10 patients, six of 10 had hypotonia, four of 10 had acquired microcephaly, and five of 10 had dysmorphic features, albeit different to those typically seen in 1q41-q42 deletion syndrome and WDR26 encephalopathy. We distinguish FBXO28 encephalopathy from both of these disorders with more severe intellectual impairment, drug-resistant epilepsy, and hyperkinetic movement disorders.


Asunto(s)
Anomalías Craneofaciales/genética , Discapacidad Intelectual/genética , Epilepsias Mioclónicas Progresivas/genética , Proteínas Ligasas SKP Cullina F-box/genética , Espasmos Infantiles/genética , Adolescente , Adulto , Encefalopatías/complicaciones , Encefalopatías/genética , Encefalopatías/fisiopatología , Niño , Preescolar , Codón sin Sentido , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/fisiopatología , Epilepsia Refractaria/complicaciones , Epilepsia Refractaria/genética , Epilepsia Refractaria/fisiopatología , Electroencefalografía , Síndromes Epilépticos/complicaciones , Síndromes Epilépticos/genética , Síndromes Epilépticos/fisiopatología , Femenino , Mutación del Sistema de Lectura , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/fisiopatología , Masculino , Mutación Missense , Epilepsias Mioclónicas Progresivas/complicaciones , Epilepsias Mioclónicas Progresivas/fisiopatología , Fenotipo , Espasmos Infantiles/complicaciones , Espasmos Infantiles/fisiopatología , Adulto Joven
6.
Hum Mol Genet ; 27(4): 589-600, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29267967

RESUMEN

FRMPD4 (FERM and PDZ Domain Containing 4) is a neural scaffolding protein that interacts with PSD-95 to positively regulate dendritic spine morphogenesis, and with mGluR1/5 and Homer to regulate mGluR1/5 signaling. We report the genetic and functional characterization of 4 FRMPD4 deleterious mutations that cause a new X-linked intellectual disability (ID) syndrome. These mutations were found to be associated with ID in ten affected male patients from four unrelated families, following an apparent X-linked mode of inheritance. Mutations include deletion of an entire coding exon, a nonsense mutation, a frame-shift mutation resulting in premature termination of translation, and a missense mutation involving a highly conserved amino acid residue neighboring FRMPD4-FERM domain. Clinical features of these patients consisted of moderate to severe ID, language delay and seizures alongside with behavioral and/or psychiatric disturbances. In-depth functional studies showed that a frame-shift mutation, FRMPD4p.Cys618ValfsX8, results in a disruption of FRMPD4 binding with PSD-95 and HOMER1, and a failure to increase spine density in transfected hippocampal neurons. Behavioral studies of frmpd4-KO mice identified hippocampus-dependent spatial learning and memory deficits in Morris Water Maze test. These findings point to an important role of FRMPD4 in normal cognitive development and function in humans and mice, and support the hypothesis that FRMPD4 mutations cause ID by disrupting dendritic spine morphogenesis in glutamatergic neurons.


Asunto(s)
Espinas Dendríticas/metabolismo , Discapacidad Intelectual/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Adolescente , Adulto , Anciano , Exones/genética , Femenino , Mutación del Sistema de Lectura/genética , Humanos , Masculino , Persona de Mediana Edad , Morfogénesis/genética , Morfogénesis/fisiología , Mutación/genética , Neurogénesis/genética , Neurogénesis/fisiología , Linaje , Adulto Joven
7.
Ann Neurol ; 84(3): 436-451, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30048010

RESUMEN

OBJECTIVE: Deletions of CACNA1A, encoding the α1 subunit of CaV 2.1 channels, cause epilepsy with ataxia in humans. Whereas the deletion of Cacna1a in γ-aminobutyric acidergic (GABAergic) interneurons (INs) derived from the medial ganglionic eminence (MGE) impairs cortical inhibition and causes generalized seizures in Nkx2.1Cre ;Cacna1ac/c mice, the targeted deletion of Cacna1a in somatostatin-expressing INs (SOM-INs), a subset of MGE-derived INs, does not result in seizures, indicating a crucial role of parvalbumin-expressing (PV) INs. Here we identify the cellular and network consequences of Cacna1a deletion specifically in PV-INs. METHODS: We generated PVCre ;Cacna1ac/c mutant mice carrying a conditional Cacna1a deletion in PV neurons and evaluated the cortical cellular and network outcomes of this mutation by combining immunohistochemical assays, in vitro electrophysiology, 2-photon imaging, and in vivo video-electroencephalographic recordings. RESULTS: PVCre ;Cacna1ac/c mice display reduced cortical perisomatic inhibition and frequent absences but only rare motor seizures. Compared to Nkx2.1Cre ;Cacna1ac/c mice, PVCre ;Cacna1ac/c mice have a net increase in cortical inhibition, with a gain of dendritic inhibition through sprouting of SOM-IN axons, largely preventing motor seizures. This beneficial compensatory remodeling of cortical GABAergic innervation is mTORC1-dependent and its inhibition with rapamycin leads to a striking increase in motor seizures. Furthermore, we show that a direct chemogenic activation of cortical SOM-INs prevents motor seizures in a model of kainate-induced seizures. INTERPRETATION: Our findings provide novel evidence suggesting that the remodeling of cortical inhibition, with an mTOR-dependent gain of dendritic inhibition, determines the seizure phenotype in generalized epilepsy and that mTOR inhibition can be detrimental in epilepsies not primarily due to mTOR hyperactivation. Ann Neurol 2018;84:436-451.


Asunto(s)
Epilepsia Generalizada/prevención & control , Interneuronas/metabolismo , Convulsiones/prevención & control , Convulsiones/fisiopatología , Animales , Corteza Cerebral/metabolismo , Epilepsia Generalizada/metabolismo , Epilepsia Generalizada/fisiopatología , Neuronas GABAérgicas/citología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Eminencia Media/citología , Ratones Transgénicos , Convulsiones/metabolismo , Ácido gamma-Aminobutírico/metabolismo
8.
Ann Neurol ; 83(6): 1089-1095, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29518281

RESUMEN

VPS13 protein family members VPS13A through VPS13C have been associated with various recessive movement disorders. We describe the first disease association of rare recessive VPS13D variants including frameshift, missense, and partial duplication mutations with a novel complex, hyperkinetic neurological disorder. The clinical features include developmental delay, a childhood onset movement disorder (chorea, dystonia, or tremor), and progressive spastic ataxia or paraparesis. Characteristic brain magnetic resonance imaging shows basal ganglia or diffuse white matter T2 hyperintensities as seen in Leigh syndrome and choreoacanthocytosis. Muscle biopsy in 1 case showed mitochondrial aggregates and lipidosis, suggesting mitochondrial dysfunction. These findings underline the importance of the VPS13 complex in neurological diseases and a possible role in mitochondrial function. Ann Neurol 2018;83:1089-1095.


Asunto(s)
Discapacidad Intelectual/genética , Trastornos del Movimiento/genética , Espasticidad Muscular/genética , Mutación/genética , Atrofia Óptica/genética , Proteínas/genética , Ataxias Espinocerebelosas/genética , Ganglios Basales/patología , Encéfalo/patología , Niño , Humanos , Enfermedad de Leigh/patología , Imagen por Resonancia Magnética/métodos , Espasticidad Muscular/patología , Linaje
9.
Epilepsia ; 60(9): 1881-1894, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31468518

RESUMEN

OBJECTIVE: Developmental epileptic encephalopathies (DEEs) are genetically heterogeneous severe childhood-onset epilepsies with developmental delay or cognitive deficits. In this study, we explored the pathogenic mechanisms of DEE-associated de novo mutations in the CACNA1A gene. METHODS: We studied the functional impact of four de novo DEE-associated CACNA1A mutations, including the previously described p.A713T variant and three novel variants (p.V1396M, p.G230V, and p.I1357S). Mutant cDNAs were expressed in HEK293 cells, and whole-cell voltage-clamp recordings were conducted to test the impacts on CaV 2.1 channel function. Channel localization and structure were assessed with immunofluorescence microscopy and three-dimensional (3D) modeling. RESULTS: We find that the G230V and I1357S mutations result in loss-of-function effects with reduced whole-cell current densities and decreased channel expression at the cell membrane. By contrast, the A713T and V1396M variants resulted in gain-of-function effects with increased whole-cell currents and facilitated current activation (hyperpolarized shift). The A713T variant also resulted in slower current decay. 3D modeling predicts conformational changes favoring channel opening for A713T and V1396M. SIGNIFICANCE: Our findings suggest that both gain-of-function and loss-of-function CACNA1A mutations are associated with similarly severe DEEs and that functional validation is required to clarify the underlying molecular mechanisms and to guide therapies.


Asunto(s)
Encefalopatías/genética , Canales de Calcio/genética , Mutación con Ganancia de Función , Síndrome de Lennox-Gastaut/genética , Mutación con Pérdida de Función , Espasmos Infantiles/genética , Animales , Células Cultivadas , Femenino , Células HEK293 , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Técnicas de Placa-Clamp , Fenotipo
10.
Am J Hum Genet ; 94(6): 891-7, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24814191

RESUMEN

Epileptic encephalopathies are increasingly thought to be of genetic origin, although the exact etiology remains uncertain in many cases. We describe here three girls from two nonconsanguineous families affected by a clinical entity characterized by dysmorphic features, early-onset intractable epilepsy, intellectual disability, and cortical blindness. In individuals from each family, brain imaging also showed specific changes, including an abnormally marked pontobulbar sulcus and abnormal signals (T2 hyperintensities) and atrophy in the occipital lobe. Exome sequencing performed in the first family did not reveal any gene with rare homozygous variants shared by both affected siblings. It did, however, show one gene, DOCK7, with two rare heterozygous variants (c.2510delA [p.Asp837Alafs(∗)48] and c.3709C>T [p.Arg1237(∗)]) found in both affected sisters. Exome sequencing performed in the proband of the second family also showed the presence of two rare heterozygous variants (c.983C>G [p.Ser328(∗)] and c.6232G>T [p.Glu2078(∗)]) in DOCK7. Sanger sequencing confirmed that all three individuals are compound heterozygotes for these truncating mutations in DOCK7. These mutations have not been observed in public SNP databases and are predicted to abolish domains critical for DOCK7 function. DOCK7 codes for a Rac guanine nucleotide exchange factor that has been implicated in the genesis and polarization of newborn pyramidal neurons and in the morphological differentiation of GABAergic interneurons in the developing cortex. All together, these observations suggest that loss of DOCK7 function causes a syndromic form of epileptic encephalopathy by affecting multiple neuronal processes.


Asunto(s)
Ceguera Cortical/genética , Epilepsia/genética , Proteínas Activadoras de GTPasa/genética , Discapacidad Intelectual/genética , Niño , Preescolar , Epilepsias Mioclónicas/genética , Exoma , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Genes Recesivos , Factores de Intercambio de Guanina Nucleótido/genética , Heterocigoto , Homocigoto , Humanos , Lactante , Masculino , Mutación , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Espasmos Infantiles/genética
11.
Hum Mol Genet ; 23(18): 4846-58, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24781210

RESUMEN

Infantile spasms (IS) is an early-onset epileptic encephalopathy of unknown etiology in ∼40% of patients. We hypothesized that unexplained IS cases represent a large collection of rare single-gene disorders. We investigated 44 children with unexplained IS using comparative genomic hybridisation arrays (aCGH) (n = 44) followed by targeted sequencing of 35 known epilepsy genes (n = 8) or whole-exome sequencing (WES) of familial trios (n = 18) to search for rare inherited or de novo mutations. aCGH analysis revealed de novo variants in 7% of patients (n = 3/44), including a distal 16p11.2 duplication, a 15q11.1q13.1 tetrasomy and a 2q21.3-q22.2 deletion. Furthermore, it identified a pathogenic maternally inherited Xp11.2 duplication. Targeted sequencing was informative for ARX (n = 1/14) and STXBP1 (n = 1/8). In contrast, sequencing of a panel of 35 known epileptic encephalopathy genes (n = 8) did not identify further mutations. Finally, WES (n = 18) was very informative, with an excess of de novo mutations identified in genes predicted to be involved in neurodevelopmental processes and/or known to be intolerant to functional variations. Several pathogenic mutations were identified, including de novo mutations in STXBP1, CASK and ALG13, as well as recessive mutations in PNPO and ADSL, together explaining 28% of cases (5/18). In addition, WES identified 1-3 de novo variants in 64% of remaining probands, pointing to several interesting candidate genes. Our results indicate that IS are genetically heterogeneous with a major contribution of de novo mutations and that WES is significantly superior to targeted re-sequencing in identifying detrimental genetic variants involved in IS.


Asunto(s)
Cromosomas Humanos/genética , Mutación , Espasmos Infantiles/genética , Preescolar , Deleción Cromosómica , Duplicación Cromosómica , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Lactante , Recién Nacido , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ADN , Espasmos Infantiles/patología , Tetrasomía
12.
Hum Mutat ; 36(8): 753-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25864427

RESUMEN

NALCN and its homologues code for the ion channel responsible for half of background Na(+) -leak conductance in vertebrate and invertebrate neurons. Recessive mutations in human NALCN cause intellectual disability (ID) with hypotonia. Here, we report a de novo heterozygous mutation in NALCN affecting a conserved residue (p.R1181Q) in a girl with ID, episodic and persistent ataxia, and arthrogryposis. Interestingly, her episodes of ataxia were abolished by the administration of acetazolamide, similar to the response observed in episodic ataxia associated with other ion channels. Introducing the analogous mutation in the Caenorhabditis elegans homologue nca-1 induced a coiling locomotion phenotype, identical to that obtained with previously characterized C. elegans gain-of-function nca alleles, suggesting that p.R1181Q confers the same property to NALCN. This observation thus suggests that dominant mutations in NALCN can cause a neurodevelopmental phenotype that overlaps with, while being mostly distinct from that associated with recessive mutations in the same gene.


Asunto(s)
Artrogriposis/genética , Ataxia/genética , Discapacidad Intelectual/genética , Mutación , Canales de Sodio/genética , Acetazolamida/uso terapéutico , Animales , Artrogriposis/metabolismo , Ataxia/tratamiento farmacológico , Ataxia/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Preescolar , Femenino , Humanos , Discapacidad Intelectual/metabolismo , Canales Iónicos/genética , Proteínas de la Membrana , Canales de Sodio/metabolismo
13.
Hum Mutat ; 36(1): 69-78, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25265257

RESUMEN

KIF1A is a neuron-specific motor protein that plays important roles in cargo transport along neurites. Recessive mutations in KIF1A were previously described in families with spastic paraparesis or sensory and autonomic neuropathy type-2. Here, we report 11 heterozygous de novo missense mutations (p.S58L, p.T99M, p.G102D, p.V144F, p.R167C, p.A202P, p.S215R, p.R216P, p.L249Q, p.E253K, and p.R316W) in KIF1A in 14 individuals, including two monozygotic twins. Two mutations (p.T99M and p.E253K) were recurrent, each being found in unrelated cases. All these de novo mutations are located in the motor domain (MD) of KIF1A. Structural modeling revealed that they alter conserved residues that are critical for the structure and function of the MD. Transfection studies suggested that at least five of these mutations affect the transport of the MD along axons. Individuals with de novo mutations in KIF1A display a phenotype characterized by cognitive impairment and variable presence of cerebellar atrophy, spastic paraparesis, optic nerve atrophy, peripheral neuropathy, and epilepsy. Our findings thus indicate that de novo missense mutations in the MD of KIF1A cause a phenotype that overlaps with, while being more severe, than that associated with recessive mutations in the same gene.


Asunto(s)
Trastornos del Conocimiento/genética , Cinesinas/química , Cinesinas/genética , Enfermedades del Sistema Nervioso/genética , Paraparesia Espástica/genética , Adolescente , Adulto , Niño , Preescolar , Trastornos del Conocimiento/patología , Epilepsia/genética , Epilepsia/patología , Neuropatías Hereditarias Sensoriales y Autónomas/genética , Neuropatías Hereditarias Sensoriales y Autónomas/patología , Humanos , Masculino , Modelos Moleculares , Mutación Missense , Enfermedades del Sistema Nervioso/patología , Paraparesia Espástica/patología , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Estructura Terciaria de Proteína , Adulto Joven
14.
Ann Neurol ; 74(2): 209-22, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23595603

RESUMEN

OBJECTIVE: Both the neuronal populations and mechanisms responsible for generalized spike-wave absence seizures are poorly understood. In mutant mice carrying loss-of-function (LOF) mutations in Cacna1a, which encodes the α1 pore-forming subunit of CaV 2.1 (P/Q-type) voltage-gated Ca(2+) channels, generalized spike-wave seizures have been suggested to result from excessive bursting of thalamocortical cells. However, other cellular populations including cortical inhibitory interneurons may contribute to this phenotype. We investigated how different cortical interneuron subtypes are affected by the loss of CaV 2.1 channel function and how this contributes to the onset of generalized epilepsy. METHODS: We designed genetic strategies to induce a selective Cacna1a LOF mutation in different cortical γ-aminobutyric acidergic (GABAergic) and/or glutamatergic neuronal populations in mice. We assessed the cellular and network consequences of these mutations by combining immunohistochemical assays, in vitro physiology, optogenetics, and in vivo video electroencephalographic recordings. RESULTS: We demonstrate that selective Cacna1a LOF from a subset of cortical interneurons, including parvalbumin (PV)(+) and somatostatin (SST)(+) interneurons, results in severe generalized epilepsy. Loss of CaV 2.1 channel function compromises GABA release from PV(+) but not SST(+) interneurons. Moreover, thalamocortical projection neurons do not show enhanced bursting in these mutants, suggesting that this feature is not essential for the development of generalized spike-wave seizures. Notably, the concurrent removal of CaV 2.1 channels in cortical pyramidal cells and interneurons considerably lessens seizure severity by decreasing cortical excitability. INTERPRETATION: Our findings demonstrate that conditional ablation of CaV 2.1 channel function from cortical PV(+) interneurons alters GABA release from these cells, impairs their ability to constrain cortical pyramidal cell excitability, and is sufficient to cause generalized seizures.


Asunto(s)
Canales de Calcio Tipo N/fisiología , Corteza Cerebral/fisiopatología , Interneuronas/patología , Convulsiones/patología , Animales , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Interneuronas/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Parvalbúminas/metabolismo , Fenotipo , Convulsiones/etiología , Convulsiones/metabolismo , Somatostatina/metabolismo , Ácido gamma-Aminobutírico/metabolismo
15.
Epilepsia ; 55(8): 1170-86, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24965021

RESUMEN

New genetic investigation techniques, including next-generation sequencing, epigenetic profiling, cell lineage mapping, targeted genetic manipulation of specific neuronal cell types, stem cell reprogramming, and optogenetic manipulations within epileptic networks are progressively unraveling the mysteries of epileptogenesis and ictogenesis. These techniques have opened new avenues to discover the molecular basis of epileptogenesis and to study the physiologic effects of mutations in epilepsy-associated genes on a multilayer level, from cells to circuits. This manuscript reviews recently published applications of these new genetic technologies in the study of epilepsy, as well as work presented by the authors at the genetic session of the XII Workshop on the Neurobiology of Epilepsy (WONOEP 2013) in Quebec, Canada. Next-generation sequencing is providing investigators with an unbiased means to assess the molecular causes of sporadic forms of epilepsy and has revealed the complexity and genetic heterogeneity of sporadic epilepsy disorders. To assess the functional impact of mutations in these newly identified genes on specific neuronal cell types during brain development, new modeling strategies in animals, including conditional genetics in mice and in utero knock-down approaches, are enabling functional validation with exquisite cell-type and temporal specificity. In addition, optogenetics, using cell-type-specific Cre recombinase driver lines, is enabling investigators to dissect networks involved in epilepsy. In addition, genetically encoded cell-type labeling is providing new means to assess the role of the nonneuronal components of epileptic networks such as glial cells. Furthermore, beyond its role in revealing coding variants involved in epileptogenesis, next-generation sequencing can be used to assess the epigenetic modifications that lead to sustained network hyperexcitability in epilepsy, including methylation changes in gene promoters and noncoding ribonucleic acid (RNA) involved in modifying gene expression following seizures. In addition, genetically based bioluminescent reporters are providing new opportunities to assess neuronal activity and neurotransmitter levels both in vitro and in vivo in the context of epilepsy. Finally, genetically rederived neurons generated from patient induced pluripotent stem cells and genetically modified zebrafish have become high-throughput means to investigate disease mechanisms and potential new therapies. Genetics has changed the field of epilepsy research considerably, and is paving the way for better diagnosis and therapies for patients with epilepsy.


Asunto(s)
Educación/métodos , Epigénesis Genética/genética , Epilepsia/diagnóstico , Epilepsia/genética , Hibridación Genética/genética , Animales , Humanos , MicroARNs/genética
16.
Muscle Nerve ; 49(1): 134-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23893323

RESUMEN

INTRODUCTION: Erythromelalgia due to heterozygous gain-of-function SCN9A mutations usually presents as a pure sensory-autonomic disorder characterized by recurrent episodes of burning pain and redness of the extremities. METHODS: We describe a patient with an unusual phenotypic presentation of gross motor delay, childhood-onset erythromelalgia, extreme visceral pain episodes, hypesthesia, and self-mutilation. The investigation of the patient's motor delay included various biochemical analyses, a comparative genomic hybridization array (CGH), electromyogram (EMG), and muscle biopsy. Once erythromelalgia was suspected clinically, the SCN9A gene was sequenced. RESULTS: The EMG, CGH, and biochemical tests were negative. The biopsy showed an axonal neuropathy and neurogenic atrophy. Sequencing of SCN9A revealed a heterozygous missense mutation in exon 7; p.I234T. CONCLUSIONS: This is a case of global motor delay and erythromelalgia associated with SCN9A. The motor delay may be attributed to the extreme pain episodes or to a developmental perturbation of proprioceptive inputs.


Asunto(s)
Trastornos de la Destreza Motora/genética , Mutación Missense/genética , Canal de Sodio Activado por Voltaje NAV1.7/genética , Índice de Severidad de la Enfermedad , Trastornos Somatomorfos/genética , Carbamazepina/uso terapéutico , Preescolar , Comorbilidad , Eritromelalgia/tratamiento farmacológico , Eritromelalgia/epidemiología , Eritromelalgia/genética , Femenino , Humanos , Hipoestesia/tratamiento farmacológico , Hipoestesia/epidemiología , Hipoestesia/genética , Mexiletine/uso terapéutico , Trastornos de la Destreza Motora/tratamiento farmacológico , Trastornos de la Destreza Motora/epidemiología , Trastornos Somatomorfos/tratamiento farmacológico , Trastornos Somatomorfos/epidemiología , Resultado del Tratamiento
17.
18.
Ther Adv Rare Dis ; 5: 26330040241245725, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38681799

RESUMEN

CACNA1A-related disorders are rare neurodevelopmental disorders linked to variants in the CACNA1A gene. This gene encodes the α1 subunit of the P/Q-type calcium channel Cav2.1, which is globally expressed in the brain and crucial for fast synaptic neurotransmission. The broad spectrum of CACNA1A-related neurological disorders includes developmental and epileptic encephalopathies, familial hemiplegic migraine type 1, episodic ataxia type 2, spinocerebellar ataxia type 6, together with unclassified presentations with developmental delay, ataxia, intellectual disability, autism spectrum disorder, and language impairment. The severity of each disorder is also highly variable. The spectrum of CACNA1A-related seizures is broad across both loss-of-function and gain-of-function variants and includes absence seizures, focal seizures with altered consciousness, generalized tonic-clonic seizures, tonic seizures, status epilepticus, and infantile spasms. Furthermore, over half of CACNA1A-related epilepsies are refractory to current therapies. To date, almost 1700 CACNA1A variants have been reported in ClinVar, with over 400 listed as Pathogenic or Likely Pathogenic, but with limited-to-no clinical or functional data. Robust genotype-phenotype studies and impacts of variants on protein structure and function have also yet to be established. As a result, there are few definitive treatment options for CACNA1A-related epilepsies. The CACNA1A Foundation has set out to change the landscape of available and effective treatments and improve the quality of life for those living with CACNA1A-related disorders, including epilepsy. Established in March 2020, the Foundation has built a robust preclinical toolbox that includes patient-derived induced pluripotent stem cells and novel disease models, launched clinical trial readiness initiatives, and organized a global CACNA1A Research Network. This Research Network is currently composed of over 60 scientists and clinicians committed to collaborating to accelerate the path to CACNA1A-specific treatments and one day, a cure.


Designing a plan to find treatments for epilepsies linked to the CACNA1A gene and test them in clinical trials for FDA approval CACNA1A-related disorders are rare conditions that affect brain development and are caused by changes in the CACNA1A gene. This gene provides instructions for making a protein called Cav2.1, which plays a crucial role in fast communication between nerve cells. The disorders can lead to various neurological problems such as seizures, epilepsy, developmental delays, intellectual disability, and autism. The severity of these disorders varies, and individuals may experience a broad range of seizures. More than 1700 different genetic changes in the CACNA1A gene have been identified, with over 400 considered likely to cause the disorders. However, there is limited information on the clinical and molecular aspects of these changes. Despite the significant impact on individuals' lives, there are currently no definitive treatments for CACNA1A-related epilepsies. To address this gap, the CACNA1A Foundation was established in March 2020. The Foundation aims to improve the lives of individuals with CACNA1A-related disorders, including epilepsy. It has developed a comprehensive set of tools, including patient-derived cells and new disease models, to advance research. Additionally, the Foundation has initiated initiatives to prepare for clinical trials and has formed a global CACNA1A Research Network with over 60 scientists and clinicians collaborating to develop specific treatments and, ultimately, find a cure.

19.
J Neurosci ; 32(49): 17690-705, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23223290

RESUMEN

Although previous work identified transcription factors crucial for the specification and migration of parvalbumin (PV)-expressing and somatostatin (SST)-expressing interneurons, the intrinsic factors required for the terminal differentiation, connectivity, and survival of these cell types remain uncharacterized. Here we demonstrate that, within subpopulations of cortical interneurons, Satb1 (special AT-rich binding protein) promotes terminal differentiation, connectivity, and survival in interneurons that express PV and SST. We find that conditional removal of Satb1 in mouse interneurons results in the loss of a majority of SST-expressing cells across all cortical layers, as well as some PV-expressing cells in layers IV and VI, by postnatal day 21. SST-expressing cells initially migrate to the cortex in Satb1 mutant mice, but receive reduced levels of afferent input and begin to die during the first postnatal week. Electrophysiological characterization indicates that loss of Satb1 function in interneurons results in a loss of functional inhibition of excitatory principal cells. These data suggest that Satb1 is required for medial ganglionic eminence-derived interneuron differentiation, connectivity, and survival.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/crecimiento & desarrollo , Interneuronas/fisiología , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Terminales Presinápticos/fisiología , Animales , Ondas Encefálicas/fisiología , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Interneuronas/citología , Interneuronas/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/fisiología , Parvalbúminas/metabolismo , Terminales Presinápticos/metabolismo , Somatostatina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
20.
Front Neural Circuits ; 17: 1256455, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37779671

RESUMEN

Cortical GABAergic interneurons are critical components of neural networks. They provide local and long-range inhibition and help coordinate network activities involved in various brain functions, including signal processing, learning, memory and adaptative responses. Disruption of cortical GABAergic interneuron migration thus induces profound deficits in neural network organization and function, and results in a variety of neurodevelopmental and neuropsychiatric disorders including epilepsy, intellectual disability, autism spectrum disorders and schizophrenia. It is thus of paramount importance to elucidate the specific mechanisms that govern the migration of interneurons to clarify some of the underlying disease mechanisms. GABAergic interneurons destined to populate the cortex arise from multipotent ventral progenitor cells located in the ganglionic eminences and pre-optic area. Post-mitotic interneurons exit their place of origin in the ventral forebrain and migrate dorsally using defined migratory streams to reach the cortical plate, which they enter through radial migration before dispersing to settle in their final laminar allocation. While migrating, cortical interneurons constantly change their morphology through the dynamic remodeling of actomyosin and microtubule cytoskeleton as they detect and integrate extracellular guidance cues generated by neuronal and non-neuronal sources distributed along their migratory routes. These processes ensure proper distribution of GABAergic interneurons across cortical areas and lamina, supporting the development of adequate network connectivity and brain function. This short review summarizes current knowledge on the cellular and molecular mechanisms controlling cortical GABAergic interneuron migration, with a focus on tangential migration, and addresses potential avenues for cell-based interneuron progenitor transplants in the treatment of neurodevelopmental disorders and epilepsy.


Asunto(s)
Corteza Cerebral , Neurogénesis , Corteza Cerebral/fisiología , Interneuronas/fisiología , Movimiento Celular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA