Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Mol Autism ; 13(1): 17, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35395866

RESUMEN

BACKGROUND: Phelan-McDermid syndrome (PMS) is caused by haploinsufficiency of the SHANK3 gene and is characterized by global developmental delays and autism spectrum disorder (ASD). Based on several converging lines of preclinical and clinical evidence supporting the use of insulin-like growth factor-1 (IGF-1) in PMS, this study aims to follow-up a previous pilot study with IGF-1 to further evaluate this novel therapeutic for core symptoms of ASD in children with PMS. METHODS: Ten children aged 5-9 with PMS were enrolled. Participants were randomized to receive IGF-1 or placebo (saline) using a 12-week, double-blind, crossover design. Efficacy was assessed using the primary outcome of the Aberrant Behavior Checklist-Social Withdrawal (ABC-SW) subscale as well as secondary outcome measures reflecting core symptoms of ASD. To increase power and sample size, we jointly analyzed the effect of IGF-1 reported here together with results from our previous controlled trail of IGF-1 in children with PMS (combined N = 19). RESULTS: Results on the ABC-SW did not reach statistical significance, however significant improvements in sensory reactivity symptoms were observed. In our pooled analyses, IGF-1 treatment also led to significant improvements in repetitive behaviors and hyperactivity. There were no other statistically significant effects seen across other clinical outcome measures. IGF-1 was well tolerated and there were no serious adverse events. LIMITATIONS: The small sample size and expectancy bias due to relying on parent reported outcome measures may contribute to limitations in interpreting results. CONCLUSION: IGF-1 is efficacious in improving sensory reactivity symptoms, repetitive behaviors, and hyperactivity  in children with PMS. Trial registration NCT01525901.


Asunto(s)
Trastornos de los Cromosomas , Factor I del Crecimiento Similar a la Insulina , Niño , Deleción Cromosómica , Trastornos de los Cromosomas/tratamiento farmacológico , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 22 , Humanos , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Proyectos Piloto
2.
Mol Autism ; 13(1): 6, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093163

RESUMEN

BACKGROUND: Phelan-McDermid syndrome (PMS) is caused by 22q13 deletions including SHANK3 or pathogenic sequence variants in SHANK3 and is among the more common rare genetic findings in autism spectrum disorder (ASD). SHANK3 is critical for synaptic function, and preclinical and clinical studies suggest that insulin-like growth factor-1 (IGF-1) can reverse a range of deficits in PMS. IGF-1 release is stimulated by growth hormone secretion from the anterior pituitary gland, and this study sought to assess the feasibility of increasing IGF-1 levels through recombinant human growth hormone (rhGH) treatment, in addition to establishing safety and exploring efficacy of rhGH in children with PMS. METHODS: rhGH was administered once daily for 12 weeks to six children with PMS using an open-label design. IGF-1 levels, safety, and efficacy assessments were measured every 4 weeks throughout the study. RESULTS: rhGH administration increased levels of IGF-1 by at least 2 standard deviations and was well tolerated without serious adverse events. rhGH treatment was also associated with clinical improvement in social withdrawal, hyperactivity, and sensory symptoms. LIMITATIONS: Results should be interpreted with caution given the small sample size and lack of a placebo control. CONCLUSIONS: Overall, findings are promising and indicate the need for larger studies with rhGH in PMS. Trial registration NCT04003207. Registered July 1, 2019, https://clinicaltrials.gov/ct2/show/NCT04003207 .


Asunto(s)
Trastorno del Espectro Autista , Hormona de Crecimiento Humana , Trastorno del Espectro Autista/genética , Niño , Deleción Cromosómica , Trastornos de los Cromosomas , Cromosomas Humanos Par 22 , Hormona del Crecimiento , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Factor I del Crecimiento Similar a la Insulina
3.
Mol Autism ; 12(1): 62, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34593045

RESUMEN

BACKGROUND: Phelan-McDermid syndrome (PMS) is a rare neurodevelopmental disorder caused by haploinsufficiency of the SHANK3 gene and characterized by global developmental delays, deficits in speech and motor function, and autism spectrum disorder (ASD). Monogenic causes of ASD such as PMS are well suited to investigations with novel therapeutics, as interventions can be targeted based on established genetic etiology. While preclinical studies have demonstrated that the neuropeptide oxytocin can reverse electrophysiological, attentional, and social recognition memory deficits in Shank3-deficient rats, there have been no trials in individuals with PMS. The purpose of this study is to assess the efficacy and safety of intranasal oxytocin as a treatment for the core symptoms of ASD in a cohort of children with PMS. METHODS: Eighteen children aged 5-17 with PMS were enrolled. Participants were randomized to receive intranasal oxytocin or placebo (intranasal saline) and underwent treatment during a 12-week double-blind, parallel group phase, followed by a 12-week open-label extension phase during which all participants received oxytocin. Efficacy was assessed using the primary outcome of the Aberrant Behavior Checklist-Social Withdrawal (ABC-SW) subscale as well as a number of secondary outcome measures related to the core symptoms of ASD. Safety was monitored throughout the study period. RESULTS: There was no statistically significant improvement with oxytocin as compared to placebo on the ABC-SW (Mann-Whitney U = 50, p = 0.055), or on any secondary outcome measures, during either the double-blind or open-label phases. Oxytocin was generally well tolerated, and there were no serious adverse events. LIMITATIONS: The small sample size, potential challenges with drug administration, and expectancy bias due to relying on parent reported outcome measures may all contribute to limitations in interpreting results. CONCLUSION: Our results suggest that intranasal oxytocin is not efficacious in improving the core symptoms of ASD in children with PMS. Trial registration NCT02710084.


Asunto(s)
Trastorno del Espectro Autista , Trastornos de los Cromosomas , Adolescente , Trastorno del Espectro Autista/genética , Niño , Preescolar , Deleción Cromosómica , Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/tratamiento farmacológico , Cromosomas Humanos Par 22 , Humanos , Oxitocina/uso terapéutico
4.
Clin Genet ; 92(4): 415-422, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28295210

RESUMEN

BACKGROUND: Array-comparative genomic hybridization (array-CGH) is a widely used technique to detect copy number variants (CNVs) associated with developmental delay/intellectual disability (DD/ID). AIMS: Identification of genomic disorders in DD/ID. MATERIALS AND METHODS: We performed a comprehensive array-CGH investigation of 1,015 consecutive cases with DD/ID and combined literature mining, genetic evidence, evolutionary constraint scores, and functional information in order to assess the pathogenicity of the CNVs. RESULTS: We identified non-benign CNVs in 29% of patients. Amongst the pathogenic variants (11%), detected with a yield consistent with the literature, we found rare genomic disorders and CNVs spanning known disease genes. We further identified and discussed 51 cases with likely pathogenic CNVs spanning novel candidate genes, including genes encoding synaptic components and/or proteins involved in corticogenesis. Additionally, we identified two deletions spanning potential Topological Associated Domain (TAD) boundaries probably affecting the regulatory landscape. DISCUSSION AND CONCLUSION: We show how phenotypic and genetic analyses of array-CGH data allow unraveling complex cases, identifying rare disease genes, and revealing unexpected position effects.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Adolescente , Adulto , Niño , Preescolar , Efectos de la Posición Cromosómica/genética , Aberraciones Cromosómicas , Hibridación Genómica Comparativa , Discapacidades del Desarrollo/patología , Femenino , Estudios de Asociación Genética , Genómica , Humanos , Lactante , Discapacidad Intelectual/patología , Masculino , Linaje , Fenotipo , Eliminación de Secuencia/genética , Adulto Joven
5.
J Autism Dev Disord ; 47(5): 1314-1322, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28168677

RESUMEN

Autism spectrum disorder (ASD) affects individuals across all racial and ethnic groups, yet rates of diagnosis are disproportionately higher for Black and Hispanic children. Caregivers of children with ASD experience significant stressors, which have been associated with parental strain, inadequate utilization of mental health services and lower quality of life. The family peer advocate (FPA) model has been utilized across service delivery systems to provide family-to-family support, facilitate engagement, and increase access to care. This study used a randomized controlled design to examine the efficacy of FPAs in a racially and ethnically diverse sample. Results demonstrate significantly increased knowledge of ASD and reduced levels of stress for caregivers who received the FPA intervention as compared to treatment as usual.


Asunto(s)
Trastorno del Espectro Autista/enfermería , Negro o Afroamericano/psicología , Cuidadores/psicología , Terapia Familiar , Hispánicos o Latinos/psicología , Padres/psicología , Estrés Psicológico/terapia , Adulto , Niño , Preescolar , Femenino , Conocimientos, Actitudes y Práctica en Salud , Humanos , Masculino , Servicios de Salud Mental , Aceptación de la Atención de Salud/psicología , Grupo Paritario , Calidad de Vida , Método Simple Ciego
6.
Mol Psychiatry ; 18(8): 889-97, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22850627

RESUMEN

Latrepirdine (Dimebon) is a pro-neurogenic, antihistaminic compound that has yielded mixed results in clinical trials of mild to moderate Alzheimer's disease, with a dramatically positive outcome in a Russian clinical trial that was unconfirmed in a replication trial in the United States. We sought to determine whether latrepirdine (LAT)-stimulated amyloid precursor protein (APP) catabolism is at least partially attributable to regulation of macroautophagy, a highly conserved protein catabolism pathway that is known to be impaired in brains of patients with Alzheimer's disease (AD). We utilized several mammalian cellular models to determine whether LAT regulates mammalian target of rapamycin (mTOR) and Atg5-dependent autophagy. Male TgCRND8 mice were chronically administered LAT prior to behavior analysis in the cued and contextual fear conditioning paradigm, as well as immunohistological and biochemical analysis of AD-related neuropathology. Treatment of cultured mammalian cells with LAT led to enhanced mTOR- and Atg5-dependent autophagy. Latrepirdine treatment of TgCRND8 transgenic mice was associated with improved learning behavior and with a reduction in accumulation of Aß42 and α-synuclein. We conclude that LAT possesses pro-autophagic properties in addition to the previously reported pro-neurogenic properties, both of which are potentially relevant to the treatment and/or prevention of neurodegenerative diseases. We suggest that elucidation of the molecular mechanism(s) underlying LAT effects on neurogenesis, autophagy and behavior might warranty the further study of LAT as a potentially viable lead compound that might yield more consistent clinical benefit following the optimization of its pro-neurogenic, pro-autophagic and/or pro-cognitive activities.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Autofagia/efectos de los fármacos , Cognición/efectos de los fármacos , Indoles/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Animales , Proteína 5 Relacionada con la Autofagia , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Indoles/uso terapéutico , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , alfa-Sinucleína/metabolismo
8.
Mol Psychiatry ; 17(4): 402-11, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21358714

RESUMEN

Autism spectrum disorders (ASDs) comprise a constellation of highly heritable neuropsychiatric disorders. Genome-wide studies of autistic individuals have implicated numerous minor risk alleles but few common variants, suggesting a complex genetic model with many contributing loci. To assess commonality of biological function among rare risk alleles, we compared functional knowledge of genes overlapping inherited structural variants in idiopathic ASD subjects relative to healthy controls. In this study we show that biological processes associated with synapse function and neurotransmission are significantly enriched, with replication, in ASD subjects versus controls. Analysis of phenotypes observed for mouse models of copy-variant genes established significant and replicated enrichment of observable phenotypes consistent with ASD behaviors. Most functional terms retained significance after excluding previously reported ASD loci. These results implicate several new variants that involve synaptic function and glutamatergic signaling processes as important contributors of ASD pathophysiology and suggest a sizable pool of additional potential ASD risk loci.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Variaciones en el Número de Copia de ADN/genética , Predisposición Genética a la Enfermedad/genética , Proteínas del Tejido Nervioso/genética , Sinapsis/genética , Transmisión Sináptica/genética , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Estudio de Asociación del Genoma Completo/métodos , Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Técnicas de Genotipaje/métodos , Técnicas de Genotipaje/psicología , Humanos , Masculino , Ratones , Fenotipo
9.
Transl Psychiatry ; 1: e8, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-22832403

RESUMEN

Schizophrenia is a serious and chronic mental disorder, in which both genetic and environmental factors have a role in the development of the disease. Neuregulin-1 (NRG1) is one of the most established genetic risk factors for schizophrenia, and disruption of NRG1 signaling has been reported in this disorder. We reported previously that NRG1/ErbB4 signaling is inhibited by receptor phosphotyrosine phosphatase-ß/ζ (RPTP ß/ζ) and that the gene encoding RPTPß/ζ (PTPRZ1) is genetically associated with schizophrenia. In this study, we examined the expression of RPTPß/ζ in the brains of patients with schizophrenia and observed increased expression of this gene. We developed mice overexpressing RPTPß/ζ (PTPRZ1-transgenic mice), which showed reduced NRG1 signaling, and molecular and cellular changes implicated in the pathogenesis of schizophrenia, including altered glutamatergic, GABAergic and dopaminergic activity, as well as delayed oligodendrocyte development. Behavioral analyses also demonstrated schizophrenia-like changes in the PTPRZ1-transgenic mice, including reduced sensory motor gating, hyperactivity and working memory deficits. Our results indicate that enhanced RPTPß/ζ signaling can contribute to schizophrenia phenotypes, and support both construct and face validity for PTPRZ1-transgenic mice as a model for multiple schizophrenia phenotypes. Furthermore, our results implicate RPTPß/ζ as a therapeutic target in schizophrenia.


Asunto(s)
Trastornos del Conocimiento/genética , Regulación Enzimológica de la Expresión Génica , Fenotipo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Esquizofrenia/genética , Regulación hacia Arriba/genética , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Esquizofrenia/enzimología , Esquizofrenia/metabolismo , Transducción de Señal/genética , Adulto Joven
10.
Mol Psychiatry ; 15(9): 954-68, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19401682

RESUMEN

Autism spectrum disorders are a group of highly heritable neurodevelopmental disorders with a complex genetic etiology. The International Molecular Genetic Study of Autism Consortium previously identified linkage loci on chromosomes 7 and 2, termed AUTS1 and AUTS5, respectively. In this study, we performed a high-density association analysis in AUTS1 and AUTS5, testing more than 3000 single nucleotide polymorphisms (SNPs) in all known genes in each region, as well as SNPs in non-genic highly conserved sequences. SNP genotype data were also used to investigate copy number variation within these regions. The study sample consisted of 127 and 126 families, showing linkage to the AUTS1 and AUTS5 regions, respectively, and 188 gender-matched controls. Further investigation of the strongest association results was conducted in an independent European family sample containing 390 affected individuals. Association and copy number variant analysis highlighted several genes that warrant further investigation, including IMMP2L and DOCK4 on chromosome 7. Evidence for the involvement of DOCK4 in autism susceptibility was supported by independent replication of association at rs2217262 and the finding of a deletion segregating in a sib-pair family.


Asunto(s)
Trastorno Autístico/genética , Cromosomas Humanos Par 2 , Cromosomas Humanos Par 7 , Endopeptidasas/genética , Proteínas Activadoras de GTPasa/genética , Adulto , Niño , Femenino , Dosificación de Gen , Predisposición Genética a la Enfermedad , Variación Genética , Genotipo , Humanos , Desequilibrio de Ligamiento , Masculino , Polimorfismo de Nucleótido Simple
11.
Mol Psychiatry ; 13(4): 385-97, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18180767

RESUMEN

Autism is a neurodevelopmental disorder with a strong genetic component, probably involving several genes. Genome screens have provided evidence of linkage to chromosome 2q31-q33, which includes the SLC25A12 gene. Association between autism and single-nucleotide polymorphisms in SLC25A12 has been reported in various studies. SLC25A12 encodes the mitochondrial aspartate/glutamate carrier functionally important in neurons with high-metabolic activity. Neuropathological findings and functional abnormalities in autism have been reported for Brodmann's area (BA) 46 and the cerebellum. We found that SLC25A12 was expressed more strongly in the post-mortem brain tissues of autistic subjects than in those of controls, in the BA46 prefrontal cortex but not in cerebellar granule cells. SLC25A12 expression was not modified in brain subregions of bipolar and schizophrenic patients. SLC25A12 was expressed in developing human neuronal tissues, including neocortical regions containing excitatory neurons and neocortical progenitors and the ganglionic eminences that generate neocortical inhibitory interneurons. At mid-gestation, when gyri and sulci start to develop, SLC25A12 molecular gradients were identified in the lateral prefrontal and ventral temporal cortex. These fetal structures generate regions with abnormal activity in autism, including the dorsolateral prefrontal cortex (BA46), the pars opercularis of the inferior frontal cortex and the fusiform gyrus. SLC25A12 overexpression or silencing in mouse embryonic cortical neurons also modified dendrite length and the mobility of dendritic mitochondria. Our findings suggest that SLC25A12 overexpression may be involved in the pathophysiology of autism, modifying neuronal networks in specific subregions, such as the dorsolateral prefrontal cortex and fusiform gyrus, at both pre- and postnatal stages.


Asunto(s)
Trastorno Autístico , Predisposición Genética a la Enfermedad , Proteínas de Transporte de Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Neuritas/fisiología , Polimorfismo de Nucleótido Simple/genética , Corteza Prefrontal/metabolismo , Regulación hacia Arriba/fisiología , Animales , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Trastorno Autístico/patología , Línea Celular Transformada , Células Cultivadas , Cromosomas Humanos Par 2 , Feto , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Humanos , Técnicas In Vitro , Desequilibrio de Ligamiento , Proteínas de Transporte de Membrana/genética , Ratones , Mitocondrias/fisiología , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Mitocondriales/genética , Neuronas/citología , Neuronas/metabolismo , Cambios Post Mortem , Corteza Prefrontal/embriología , Corteza Prefrontal/patología , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , ARN Mensajero/metabolismo , Telencéfalo/citología , Transfección
12.
Mol Psychiatry ; 13(2): 162-72, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17579610

RESUMEN

Neuregulin and the neuregulin receptor ERBB4 have been genetically and functionally implicated in schizophrenia. In this study, we used the yeast two-hybrid system to identify proteins that interact with ERBB4, to identify genes and pathways that might contribute to schizophrenia susceptibility. We identified the MAGI scaffolding proteins as ERBB4-binding proteins. After validating the interaction of MAGI proteins with ERBB4 in mammalian cells, we demonstrated that ERBB4 expression, alone or in combination with ERBB2 or ERBB3, led to the tyrosine phosphorylation of MAGI proteins, and that this could be further enhanced with receptor activation by neuregulin. As MAGI proteins were previously shown to interact with receptor phosphotyrosine phosphatase beta/zeta (RPTPbeta), we postulated that simultaneous binding of MAGI proteins to RPTPbeta and ERBB4 forms a phosphotyrosine kinase/phosphotyrosine phosphatase complex. Studies in cultured cells confirmed both a spatial and functional association between ERBB4, MAGI and RPTPbeta. Given the evidence for this functional association, we examined the genes coding for MAGI and RPTPbeta for genetic association with schizophrenia in a Caucasian United Kingdom case-control cohort (n= approximately 1400). PTPRZ1, which codes for RPTPbeta, showed significant, gene-wide and hypothesis-wide association with schizophrenia in our study (best individual single-nucleotide polymorphism allelic P=0.0003; gene-wide P=0.0064; hypothesis-wide P=0.026). The data provide evidence for a role of PTPRZ1, and for RPTPbeta signaling abnormalities, in the etiology of schizophrenia. Furthermore, the data indicate a role for RPTPbeta in the modulation of ERBB4 signaling that may in turn provide further support for an important role of neuregulin/ERBB4 signaling in the molecular basis of schizophrenia.


Asunto(s)
Receptores ErbB/metabolismo , Predisposición Genética a la Enfermedad , Proteínas del Tejido Nervioso/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Esquizofrenia/genética , Transducción de Señal/fisiología , Estudios de Casos y Controles , Línea Celular Tumoral , Distribución de Chi-Cuadrado , Receptores ErbB/genética , Femenino , Glioma , Humanos , Inmunoprecipitación/métodos , Masculino , Modelos Moleculares , Proteínas del Tejido Nervioso/genética , Neurregulina-1 , Polimorfismo de Nucleótido Simple , Proteínas Tirosina Fosfatasas , Receptor ErbB-4 , Transfección , Tirosina/metabolismo
13.
Neurobiol Aging ; 28(1): 1-7, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16343696

RESUMEN

The degree to which neurofibrillary tangles (NFT), the hallmark lesions of Alzheimer disease (AD), contribute to the development of the cognitive symptoms of AD has been debated. NFTs are comprised of abnormally phosphorylated and conformationally altered tau proteins. Conformational changes in tau have been proposed to be among the earliest neurobiological changes in AD. This study examined whether conformational changes detected by antibodies MC1 and TG3 represent early abnormalities in the disease process by assessing their presence at different stages of dementia in multiple brain regions. Postmortem specimens from several neocortical regions were examined for conformational changes in tau by ELISA in subjects [n=81] who died at different stages of cognitive impairment. Concentrations of conformationally altered tau increased with increasing dementia severity and the levels of MC1 immunoreactivity increased in the frontal cortex of mildly demented subjects before the appearance of NFT bearing neurons, suggesting that conformational alterations in tau occur early in the course of AD and its cognitive symptoms and may precede histologically identified NFTs.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Química Encefálica , Encéfalo/patología , Proteínas tau/química , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Proteínas tau/clasificación
14.
Mol Cell Neurosci ; 25(4): 642-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15080893

RESUMEN

BACE is an aspartyl protease that cleaves the amyloid precursor protein (APP) at the beta-secretase cleavage site and is involved in Alzheimer's disease. The aim of our study was to determine whether BACE affects the processing of the APP homolog APLP2. To this end, we developed BACE knockout mice with a targeted insertion of the gene for beta-galactosidase. BACE appeared to be exclusively expressed in neurons as determined by differential staining. BACE was expressed in specific areas in the cortex, hippocampus, cerebellum, pons, and spinal cord. APP processing was altered in the BACE knockouts with Abeta levels decreasing. The levels of APLP2 proteolytic products were decreased in BACE KO mice, but increased in BACE transgenic mice. Overexpression of BACE in cultured cells led to increased APLP2 processing. Our results strongly suggest that BACE is a neuronal protein that modulates the processing of both APP and APLP2.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Química Encefálica/genética , Encéfalo/enzimología , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/deficiencia , Ácido Aspártico Endopeptidasas/genética , Encéfalo/patología , Encéfalo/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Endopeptidasas , Genes Reporteros/genética , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/enzimología , Neuronas/patología , Transfección , beta-Galactosidasa/genética
15.
Mol Psychiatry ; 9(2): 144-50, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14699429

RESUMEN

Although there is considerable evidence for a strong genetic component to idiopathic autism, several genome-wide screens for susceptibility genes have been carried out with limited concordance of linked loci, reflecting numerous genes of weak effect and/or sample heterogeneity. In the current study, linkage analysis was carried out in a sample of 62 autism-affected relative pairs with more severe obsessive-compulsive behaviors, selected from a larger (n=115) set of autism-affected relative pairs as a means of reducing sample heterogeneity. Obsessive-compulsive behaviors were assessed using the Autism Diagnostic Interview-Revised (ADI-R). In the sample with more severe obsessive-compulsive behaviors, multipoint NPL scores above 2 were observed on chromosomes 1, 4, 5, 6, 10, 11 and 19, with the strongest evidence for linkage on chromosome 1 at the marker D1S1656, where the multipoint NPL score was 3.06, and the two-point NPL score was 3.21. In follow-up analyses, analyzing the subset of families (n=35) where the patients had the most severe obsessive-compulsive behaviors generated a multipoint NPL score of 2.76, and a two-point NPL score of 2.79, indicating that the bulk of evidence for linkage was derived from the families most severely affected with obsessive-compulsive behaviors. The data suggest that there is an autism susceptibility gene on chromosome 1 and provide further support for the presence of autism susceptibility genes on chromosomes 6 and 19.


Asunto(s)
Trastorno Autístico/genética , Cromosomas Humanos , Ligamiento Genético , Trastorno Obsesivo Compulsivo/genética , Cromosomas Humanos Par 1 , Cromosomas Humanos Par 19 , Cromosomas Humanos Par 6 , Salud de la Familia , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos
16.
Mol Cell Neurosci ; 23(3): 495-506, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12837631

RESUMEN

Calsenilin is a member of the neuronal calcium sensor (NCS) family of proteins that interacts with the presenilins. Calsenilin has been found to act as a Kv4alpha channel interactor and as a transcriptional repressor. We have recently shown that calsenilin can be cleaved by caspase-3 and that its cleavage separates the conserved calcium-binding domain from the variable N-terminal domain. Here, we demonstrate that calsenilin can be phosphorylated by casein kinase I and that its phosphorylation can be regulated by intracellular calcium. In addition, phosphorylated calsenilin is a substrate for serine/threonine protein phosphatase (PP) 1 and/or 2A. Phosphorylation within the N-terminal domain at Ser63, the major phosphorylation site of calsenilin, inhibits cleavage of the molecule by caspase-3. Given that the N-terminal domain of calsenilin is not conserved in the larger NCS family including other KChIP/CALP proteins, phosphorylation of calsenilin may regulate a functional role that is unique to this member of the superfamily.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Caspasas/metabolismo , Neuronas/enzimología , Proteínas Represoras , Animales , Especificidad de Anticuerpos , Calcio/metabolismo , Proteínas de Unión al Calcio/inmunología , Caseína Quinasas , Caspasa 3 , Humanos , Proteínas de Interacción con los Canales Kv , Ratones , Neuroblastoma , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Serina/metabolismo , Células Tumorales Cultivadas
17.
Mol Psychiatry ; 8(2): 186-94, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12610651

RESUMEN

Autism is a psychiatric disorder with estimated heritability of 90%. One-third of autistic individuals experience seizures. A susceptibility locus for autism was mapped near a cluster of voltage-gated sodium channel genes on chromosome 2. Mutations in two of these genes, SCN1A and SCN2A, result in the seizure disorder GEFS+. To evaluate these sodium channel genes as candidates for the autism susceptibility locus, we screened for variation in coding exons and splice sites in 117 multiplex autism families. A total of 27 kb of coding sequence and 3 kb of intron sequence were screened. Only six families carried variants with potential effects on sodium channel function. Five coding variants and one lariat branchpoint mutation were each observed in a single family, but were not present in controls. The variant R1902C in SCN2A is located in the calmodulin binding site and was found to reduce binding affinity for calcium-bound calmodulin. R542Q in SCN1A was observed in one autism family and had previously been identified in a patient with juvenile myoclonic epilepsy. The effect of the lariat branchpoint mutation was tested in cultured lymphoblasts. Additional population studies and functional tests will be required to evaluate pathogenicity of the coding and lariat site variants. SNP density was 1/kb in the genomic sequence screened. We report 38 sodium channel SNPs that will be useful in future association and linkage studies.


Asunto(s)
Trastorno Autístico/genética , Proteínas del Tejido Nervioso/genética , Canales de Sodio/genética , Secuencia de Bases , Línea Celular , Exones , Salud de la Familia , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Intrones , Masculino , Canal de Sodio Activado por Voltaje NAV1.1 , Canal de Sodio Activado por Voltaje NAV1.2 , Canal de Sodio Activado por Voltaje NAV1.3 , Proteínas del Tejido Nervioso/química , Linaje , Estructura Terciaria de Proteína , Canales de Sodio/química
18.
Neuroscience ; 114(1): 247-63, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12207970

RESUMEN

Mutations in the presenilin 1 and 2 genes cause the majority of early onset familial forms of Alzheimer's disease. Here we describe the biochemical and immunohistochemical characterization of calsenilin, a novel calcium binding protein that we have previously shown to interact with presenilins 1 and 2, in mouse brain. The co-immunoprecipitation of endogenous calsenilin and presenilin 1 demonstrates that these proteins are physiologic binding partners. Although calsenilin has been predicted to be a soluble protein, we have found that the majority of it is tightly associated with the cytoplasmic face of intracellular membranes and that it can only be dissociated using harsh treatments such as urea. In addition, we have demonstrated that calsenilin is a developmentally regulated protein that is mainly present in the brain, where it localizes to both the hippocampus and cerebellum. Calsenilin staining co-localized with the somatodendritic marker microtubule-associated protein-2 primarily in the granular cell layer of the cerebellum, indicating that calsenilin expression is primarily neuronal. In primary cultured neurons, calsenilin immunoreactivity was observed in cell bodies as well as in some neuronal processes. Co-localization experiments using specific axonal and dendritic markers indicate that these processes were mainly axonal in nature, although a smaller subset of dendrites also appears to contain calsenilin. In summary, we have established that calsenilin and presenilin 1 can interact at physiologic levels, and that calsenilin is a developmentally regulated protein that is expressed primarily in the cerebellum and hippocampus. Although calsenilin is a soluble protein, it is tightly associated with the membrane. Finally, the expression pattern of calsenilin, which is similar to that of the presenilin(s), suggests that the common locations of these two proteins provide an opportunity for physical interaction in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/aislamiento & purificación , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Proteínas Represoras , Envejecimiento/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Animales Recién Nacidos , Especificidad de Anticuerpos/inmunología , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Compartimento Celular/fisiología , Membrana Celular/metabolismo , Células Cultivadas , Feto , Inmunohistoquímica , Proteínas de Interacción con los Canales Kv , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Datos de Secuencia Molecular , Neuronas/citología , Presenilina-1 , Homología de Secuencia de Aminoácido
19.
Mol Cell Neurosci ; 19(4): 552-9, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11988022

RESUMEN

Calsenilin (also called DREAM and KChIP3), a member of the neuronal calcium sensor family, was isolated in a yeast two-hybrid screen using an apoptotic domain of presenilin 2 as bait. Calsenilin is a cytoplasmic protein, but interacts with the COOH-termini of both presenilin 1 and presenilin 2 at the endoplasmic reticulum and the Golgi apparatus. In this study, we have investigated calsenilin's effect on apoptosis. In stable neuroglioma cell lines, we observed that calsenilin enhances apoptosis in response to serum withdrawal or thapsigargin. Consistent with these observations, caspase and apparently calpain activities were increased during apoptosis in calsenilin-overexpressing cells. Moreover, using calcium imaging we were able to show that cells treated with thapsigargin released more calcium from intracellular stores when calsenilin was overexpressed. Taken together, these data suggest that calsenilin causes cells to be more susceptible to apoptotic triggers, possibly by altering calcium dynamics.


Asunto(s)
Apoptosis/fisiología , Señalización del Calcio/fisiología , Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Sistema Nervioso Central/metabolismo , Retículo Endoplásmico/metabolismo , Neuronas/metabolismo , Proteínas Represoras , Apoptosis/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/farmacología , Calpaína/efectos de los fármacos , Calpaína/metabolismo , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Sistema Nervioso Central/citología , Medio de Cultivo Libre de Suero/farmacología , Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas de Interacción con los Canales Kv , Neuronas/efectos de los fármacos , Tapsigargina/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
20.
Mol Psychiatry ; 7(3): 311-6, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11920158

RESUMEN

Autistic disorder (OMIM 209850) is a disease with a significant genetic component of a complex nature.(1) Cytogenetic abnormalities in the Prader-Willi/Angelman syndrome critical region (15q11-13) have been described in several individuals with autism.(1) For this reason, markers across this region have been screened for evidence of linkage and association, and a marker (155CA-2) in the gamma-aminobutyric acid type-A receptor beta3 subunit gene (GABRB3) has been associated in one study(2) but not others.(3-5) We completed an association analysis with 155CA-2 using the transmission disequilibrium test (TDT) in a set of 80 autism families (59 multiplex and 21 trios). We also used four additional markers (69CA, 155CA-1, 85CA, and A55CA-1) localized within 150 kb of 155CA-2. The use of multi-allelic TDT (MTDT) (P < 0.002), as well as the TDT (P < 0.004), demonstrated an association between autistic disorder and 155CA-2 in these families. Meiotic segregation distortion could be excluded as a possible cause for these results since no disequilibrium was observed in unaffected siblings. These findings support a role for genetic variants within the GABA receptor gene complex in 15q11-13 in autistic disorder.


Asunto(s)
Trastorno Autístico/genética , Cromosomas Humanos Par 15 , Polimorfismo Genético , Receptores de GABA-A/genética , Mapeo Cromosómico , Familia , Femenino , Marcadores Genéticos , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...