Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 370(6): 533-42, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24499211

RESUMEN

BACKGROUND: Congenital disorders of glycosylation are genetic syndromes that result in impaired glycoprotein production. We evaluated patients who had a novel recessive disorder of glycosylation, with a range of clinical manifestations that included hepatopathy, bifid uvula, malignant hyperthermia, hypogonadotropic hypogonadism, growth retardation, hypoglycemia, myopathy, dilated cardiomyopathy, and cardiac arrest. METHODS: Homozygosity mapping followed by whole-exome sequencing was used to identify a mutation in the gene for phosphoglucomutase 1 (PGM1) in two siblings. Sequencing identified additional mutations in 15 other families. Phosphoglucomutase 1 enzyme activity was assayed on cell extracts. Analyses of glycosylation efficiency and quantitative studies of sugar metabolites were performed. Galactose supplementation in fibroblast cultures and dietary supplementation in the patients were studied to determine the effect on glycosylation. RESULTS: Phosphoglucomutase 1 enzyme activity was markedly diminished in all patients. Mass spectrometry of transferrin showed a loss of complete N-glycans and the presence of truncated glycans lacking galactose. Fibroblasts supplemented with galactose showed restoration of protein glycosylation and no evidence of glycogen accumulation. Dietary supplementation with galactose in six patients resulted in changes suggestive of clinical improvement. A new screening test showed good discrimination between patients and controls. CONCLUSIONS: Phosphoglucomutase 1 deficiency, previously identified as a glycogenosis, is also a congenital disorder of glycosylation. Supplementation with galactose leads to biochemical improvement in indexes of glycosylation in cells and patients, and supplementation with complex carbohydrates stabilizes blood glucose. A new screening test has been developed but has not yet been validated. (Funded by the Netherlands Organization for Scientific Research and others.).


Asunto(s)
Glucofosfatos/genética , Enfermedad del Almacenamiento de Glucógeno/genética , Fenotipo , Fosfoglucomutasa/genética , Galactosa/uso terapéutico , Genes Recesivos , Glucosa/metabolismo , Glucofosfatos/metabolismo , Enfermedad del Almacenamiento de Glucógeno/dietoterapia , Enfermedad del Almacenamiento de Glucógeno/metabolismo , Glicoproteínas/biosíntesis , Glicosilación , Humanos , Masculino , Mutación , Fosfoglucomutasa/metabolismo , ARN Mensajero/análisis
2.
Mol Ther ; 24(6): 1030-1041, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27039844

RESUMEN

Aspartoacylase (AspA) gene mutations cause the pediatric lethal neurodegenerative Canavan disease (CD). There is emerging promise of successful gene therapy for CD using recombinant adeno-associated viruses (rAAVs). Here, we report an intracerebroventricularly delivered AspA gene therapy regime using three serotypes of rAAVs at a 20-fold reduced dose than previously described in AspA(-/-) mice, a bona-fide mouse model of CD. Interestingly, central nervous system (CNS)-restricted therapy prolonged survival over systemic therapy in CD mice but failed to sustain motor functions seen in systemically treated mice. Importantly, we reveal through histological and functional examination of untreated CD mice that AspA deficiency in peripheral tissues causes morphological and functional abnormalities in this heretofore CNS-defined disorder. We demonstrate for the first time that AspA deficiency, possibly through excessive N-acetyl aspartic acid accumulation, elicits both a peripheral and CNS immune response in CD mice. Our data establish a role for peripheral tissues in CD pathology and serve to aid the development of more efficacious and sustained gene therapy for this disease.


Asunto(s)
Amidohidrolasas/genética , Enfermedad de Canavan/terapia , Sistema Nervioso Central/patología , Terapia Genética/métodos , Animales , Enfermedad de Canavan/genética , Enfermedad de Canavan/patología , Sistema Nervioso Central/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Especificidad de Órganos , Análisis de Supervivencia , Resultado del Tratamiento
3.
Am J Med Genet A ; 167A(4): 791-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25820398

RESUMEN

Persistent müllerian duct syndrome (PMD) with antimüllerian hormone (AMH) deficiency is usually associated with mutations or deletions of the AMH gene, although many cases have no identified gene association. We report on a genetic male with PMD and AMH deficiency associated with distal monosomy 10q. A term 3,230 g infant was born to a healthy 27-year-old. Fetal ultrasound had shown possible genital ambiguity. Postnatal exam showed a 0.5 cm phallus with basal meatus, normal scrotum with no palpable gonads, no vaginal orifice, and a rectal fistula with an imperforate anus. Voiding cystourethrogram with ultrasound, cystoscopy, and laparoscopy showed normal bladder, urethral orifice, distal vagina, cervix, and bilateral abdominal testis. At 24 hours of life, testosterone was within normal range with low AMH level. Chromosome microarray analysis showed 46, XY, del10(10q25.3q26.13) involving an 8.2 MB interstitial deletion. Whole exome sequencing identified a NOTCH2 variant (1p11.2). AMH sequencing revealed no abnormalities. Following multidisciplinary team and parent discussion, male gender was assigned. Testosterone treatment resulted in penile length of 1.5 cm. Bilateral orchiopexy and posterior sagittal anorectoplasty were performed at 11 months of age; rudimentary müllerian structures were identified. This observation suggests an association of 10qter elements with male differentiation including AMH expression and is similar to a patient with 46, XY, del(10q26.1) in which AMH levels were not reported. Regional candidate genes include FGFR2 (10q26.13). The possible contribution of a NOTCH2 variant cannot be excluded.


Asunto(s)
Deleción Cromosómica , Trastorno del Desarrollo Sexual 46,XY/diagnóstico , Hormona Antimülleriana/deficiencia , Cromosomas Humanos Par 10 , Trastorno del Desarrollo Sexual 46,XY/genética , Humanos , Lactante , Masculino , Conductos Paramesonéfricos/patología
4.
Mol Ther ; 21(12): 2136-47, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23817205

RESUMEN

Canavan's disease (CD) is a fatal pediatric leukodystrophy caused by mutations in aspartoacylase (AspA) gene. Currently, there is no effective treatment for CD; however, gene therapy is an attractive approach to ameliorate the disease. Here, we studied progressive neuropathology and gene therapy in short-lived (≤ 1 month) AspA(-/-) mice, a bona-fide animal model for the severest form of CD. Single intravenous (IV) injections of several primate-derived recombinant adeno-associated viruses (rAAVs) as late as postnatal day 20 (P20) completely rescued their early lethality and alleviated the major disease symptoms, extending survival in P0-injected rAAV9 and rAAVrh8 groups to as long as 2 years thus far. We successfully used microRNA (miRNA)-mediated post-transcriptional detargeting for the first time to restrict therapeutic rAAV expression in the central nervous system (CNS) and minimize potentially deleterious effects of transgene overexpression in peripheral tissues. rAAV treatment globally improved CNS myelination, although some abnormalities persisted in the content and distribution of myelin-specific and -enriched lipids. We demonstrate that systemically delivered and CNS-restricted rAAVs can serve as efficacious and sustained gene therapeutics in a model of a severe neurodegenerative disorder even when administered as late as P20.


Asunto(s)
Amidohidrolasas/genética , Enfermedad de Canavan/terapia , Sistema Nervioso Central/patología , Dependovirus/genética , Amidohidrolasas/deficiencia , Amidohidrolasas/metabolismo , Animales , Animales Recién Nacidos , Enfermedad de Canavan/patología , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Terapia Genética , Vectores Genéticos , Humanos , Inyecciones Intravenosas , Ratones , MicroARNs/genética , Especificidad de Órganos , Difracción de Rayos X
5.
iScience ; 25(6): 104391, 2022 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-35637731

RESUMEN

Canavan disease (CD) is a devastating neurological disease that lacks effective therapy. Because CD is caused by mutations of the aspartoacylase (ASPA) gene, we introduced the wild-type (WT) ASPA gene into patient iPSCs through lentiviral transduction or CRISPR/Cas9-mediated gene editing. We then differentiated the WT ASPA-expressing patient iPSCs (ASPA-CD iPSCs) into NPCs and showed that the resultant ASPA-CD NPCs exhibited potent ASPA enzymatic activity. The ASPA-CD NPCs were able to survive in brains of transplanted CD mice. The engrafted ASPA-CD NPCs reconstituted ASPA activity in CD mouse brains, reduced the abnormally elevated level of NAA in both brain tissues and cerebrospinal fluid (CSF), and rescued hallmark pathological phenotypes of the disease, including spongy degeneration, myelination defects, and motor function impairment in transplanted CD mice. These genetically modified patient iPSC-derived NPCs represent a promising cell therapy candidate for CD, a disease that has neither a cure nor a standard treatment.

6.
Mol Genet Metab ; 102(2): 176-80, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21095151

RESUMEN

Canavan disease is a fatal neurological disease without any effective treatments to slow the relentless progress of this disorder. Enzyme replacement therapy has been used effectively to treat a number of metabolic disorders, but the presence of the blood-brain-barrier presents an additional challenge in the treatment of neurological disorders. Studies have begun with the aim of establishing a treatment protocol that can effectively replace the defective enzyme in Canavan disease patients. The human enzyme, aspartoacylase, has been cloned, expressed and purified, and the surface lysyl groups modified through PEGylation. Fully active modified enzymes were administered to mice that are defective in this enzyme and that show many of the symptoms of Canavan disease. Statistically significant increases in brain enzyme activity levels have been achieved in this animal model, as well as decreases in the elevated substrate levels that mimic those found in Canavan disease patients. These results demonstrate that the modified enzyme is gaining access to the brain and functions to correct this metabolic defect. The stage is now set for a long term study to optimize this enzyme replacement approach for the development of a treatment protocol.


Asunto(s)
Amidohidrolasas/química , Amidohidrolasas/uso terapéutico , Enfermedad de Canavan/terapia , Terapia de Reemplazo Enzimático , Animales , Enfermedad de Canavan/enzimología , Modelos Animales de Enfermedad , Portadores de Fármacos/farmacología , Estabilidad de Enzimas/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Moleculares , Polietilenglicoles/farmacología , Estructura Terciaria de Proteína , Resultado del Tratamiento
7.
Neurobiol Dis ; 40(2): 432-43, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20637282

RESUMEN

Canavan disease (CD) is a neurodegenerative disease, caused by a deficiency in the enzyme aspartoacylase (ASPA). This enzyme has been localized to oligodendrocytes; however, it is still undefined how ASPA deficiency affects oligodendrocyte development. In normal mice the pattern of ASPA expression coincides with oligodendrocyte maturation. Therefore, postnatal oligodendrocyte maturation was analyzed in ASPA-deficient mice (CD mice). Early in development, CD mice brains showed decreased expression of neural cell markers that was later compensated. In addition, the levels of myelin proteins were decreased along with abnormal myelination in CD mice compared to wild-type (WT). These defects were associated with increased global levels of acetylated histone H3, decreased chromatin compaction and increased GFAP protein, a marker for astrogliosis. Together, these findings strongly suggest that, early in postnatal development, ASPA deficiency affects oligodendrocyte maturation and myelination.


Asunto(s)
Amidohidrolasas/deficiencia , Encéfalo/patología , Enfermedad de Canavan/metabolismo , Enfermedad de Canavan/patología , Vaina de Mielina/patología , Oligodendroglía/patología , Amidohidrolasas/genética , Animales , Biomarcadores/metabolismo , Western Blotting , Encéfalo/metabolismo , Encéfalo/ultraestructura , Cromatina/metabolismo , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Histonas/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Electrónica , Vaina de Mielina/metabolismo , Vaina de Mielina/ultraestructura , Oligodendroglía/metabolismo , Oligodendroglía/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
8.
Eur J Neurosci ; 32(4): 560-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20646061

RESUMEN

The NIPSNAP (4-nitrophenylphosphatase domain and non-neuronal SNAP25-like protein homolog 1) proteins belong to a highly conserved family of proteins of unknown function. We found that NIPSNAP1 binds to the branched-chain alpha-keto acid (BCKA) dehydrogenase enzyme complex, which is disrupted in maple syrup urine disease, a disease of branched-chain amino acid catabolism that results in neurological dysfunction. Phenylketonuric (PKU) and epileptic mice show altered expression of NIPSNAP1 in the brain. Therefore, the distribution and localization of NIPSNAP1 in rat brain was determined. Results show that NIPSNAP1 is expressed exclusively in neurons including pyramidal neurons in the cerebral cortex, Purkinje neurons in the cerebellum and motor neurons in the spinal cord. Dopaminergic neurons in midbrain and noradrenergic neurons in the brainstem, which are affected in PKU, also express NIPSNAP1. NIPSNAP1 is found to be localized in the mitochondrial matrix and can bind dihydrolipoyl-transacylase and -transacetylase components of the BCKA and pyruvate dehydrogenase complexes in vitro. Our data provide the first experimental evidence for a strictly neuronal expression of this mitochondrial protein in the rat nervous system.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/metabolismo , Neuronas/metabolismo , Proteínas/metabolismo , Animales , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Femenino , Péptidos y Proteínas de Señalización Intercelular , Masculino , Enfermedad de la Orina de Jarabe de Arce/metabolismo , Proteínas de la Membrana , Ratones , Ratones Mutantes , Mitocondrias/enzimología , Mitocondrias/ultraestructura , Proteínas Mitocondriales/genética , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/anatomía & histología , Neuronas/citología , Fenilcetonurias/metabolismo , Proteínas/genética , Ratas , Ratas Sprague-Dawley
9.
Adv Sci (Weinh) ; 7(23): 2002155, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33304759

RESUMEN

Canavan disease (CD) is a fatal leukodystrophy caused by mutation of the aspartoacylase (ASPA) gene, which leads to deficiency in ASPA activity, accumulation of the substrate N-acetyl-L-aspartate (NAA), demyelination, and spongy degeneration of the brain. There is neither a cure nor a standard treatment for this disease. In this study, human induced pluripotent stem cell (iPSC)-based cell therapy is developed for CD. A functional ASPA gene is introduced into patient iPSC-derived neural progenitor cells (iNPCs) or oligodendrocyte progenitor cells (iOPCs) via lentiviral transduction or TALEN-mediated genetic engineering to generate ASPA iNPC or ASPA iOPC. After stereotactic transplantation into a CD (Nur7) mouse model, the engrafted cells are able to rescue major pathological features of CD, including deficient ASPA activity, elevated NAA levels, extensive vacuolation, defective myelination, and motor function deficits, in a robust and sustainable manner. Moreover, the transplanted mice exhibit much prolonged survival. These genetically engineered patient iPSC-derived cellular products are promising cell therapies for CD. This study has the potential to bring effective cell therapies, for the first time, to Canavan disease children who have no treatment options. The approach established in this study can also benefit many other children who have deadly genetic diseases that have no cure.

10.
Clin Infect Dis ; 49(2): 257-61, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19522649

RESUMEN

BACKGROUND: We previously reported an association between tumor necrosis factor alpha (TNFalpha)(-308)and interleukin (IL)-6(-174) polymorphisms and otitis susceptibility by history. Acute otitis media occurs most commonly as a complication of upper respiratory tract infection (URI); it is not clear why some children develop acute otitis media after URI and others do not. Our objective was to prospectively evaluate the association of TNFalpha(-308)and IL-6(-174) polymorphisms with URI and with acute otitis media development after URI. METHODS: Children aged 6-35 months were prospectively followed for occurrences of URI and acute otitis media. Blood or buccal mucosa samples were collected for DNA extraction to determine cytokine genotypes. Active and passive surveillance was used to capture all URI episodes during the 1-year follow-up period in order to study the rate of acute otitis media following URI. Data were analyzed using SAS software (SAS Institute) and general estimating equations modeling. RESULTS: Two hundred forty-two children were followed over 2689 patient-months and had DNA genotyped; 1235 URI episodes occurred, and 392 (32%) were complicated by acute otitis media. Children who had IL-6(-174) polymorphism had a higher susceptibility to URI during the study period (incidence density ratio, 1.24) and were more likely to meet established otitis susceptibility criteria (P < .01). Presence of TNFalpha(-308) polymorphism was associated with increased risk for acute otitis media after an episode of URI (odds ratio, 1.43). CONCLUSIONS: TNFalpha(-308) and IL-6(-174) genotypes are associated with increased risk for symptomatic URI and acute otitis media following URI. Future studies may be designed to carefully look at the interaction of these genetic polymorphisms with modifiable environmental risk factors.


Asunto(s)
Interleucina-6/genética , Otitis Media/epidemiología , Otitis Media/inmunología , Polimorfismo Genético , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/inmunología , Factor de Necrosis Tumoral alfa/genética , Preescolar , Susceptibilidad a Enfermedades , Femenino , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Estudios Prospectivos , Análisis de Secuencia de ADN
11.
J Neurosci Res ; 87(15): 3415-27, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19739253

RESUMEN

Loss of the oligodendrocyte (OL)-specific enzyme aspartoacylase (ASPA) from gene mutation results in the sponginess and loss of white matter (WM) in Canavan disease (CD). This study addresses the fate of OLs during the pathophysiology of CD in an adult ASPA knockout (KO) mouse strain. Massive arrays of neural stem/progenitor cells, immunopositive for PSA-NCAM, nestin, vimentin, and NG2, were observed within the severely affected spongy WM of the KO mouse brain. In these mice, G1-->S cell cycle progression was confirmed by an increase in cdk2-kinase activity, a reduction in mitotic inhibitors p21(Cip1) and p27(Kip1), and an increase in bromodeoxyuridine (BrdU) incorporation. Highly acetylated nuclear histones H2B and H3 were detected in adult KO mouse WM, suggesting the existence of noncompact chromatin as seen during early development. Costaining for BrdU- or Ki67-positive cells with markers for neural progenitors confirmed a continuous generation of OL lineage cells in KO WM. We observed a severe reduction in 21.5- and 18.5-kDa myelin basic protein and PLP/DM20 proteolipid proteins combined with a decrease in myelinated fibers and a perinuclear retention of myelin protein staining, indicating impairment in protein trafficking. Death of OLs, neurons, and astrocytes was identified in every region of the KO brain. Immature OLs constituted the largest population of dying cells, particularly in WM. We also report an early expression of full-length ASPA mRNA in normal mouse brain at embryonic day 12.5, when OL progenitors first appear during development. These findings support involvement of ASPA in CNS development and function.


Asunto(s)
Amidohidrolasas/genética , Encéfalo/anomalías , Encéfalo/enzimología , Enfermedad de Canavan/enzimología , Oligodendroglía/enzimología , Células Madre/enzimología , Animales , Biomarcadores/metabolismo , Encéfalo/fisiopatología , Enfermedad de Canavan/genética , Enfermedad de Canavan/fisiopatología , Ciclo Celular/genética , Muerte Celular/genética , Diferenciación Celular/genética , Supervivencia Celular/genética , Quinasa 2 Dependiente de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/genética , Histonas/metabolismo , Ratones , Ratones Noqueados , Proteínas de la Mielina/metabolismo , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Oligodendroglía/patología , Transporte de Proteínas/genética , ARN Mensajero/metabolismo , Células Madre/patología
12.
Genet Test ; 11(2): 174-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17627389

RESUMEN

Tetrahydrobiopterin (BH4) is a co-factor that enhances the activity of other enzymes, and this co-factor level is found to be affected in phenylketonuria (PKU), an amino acid metabolism disorder. The present study was aimed at understanding the effect of BH4 on mutations in the regulatory domain of phenylalanine hydroxylase (PAH). Among 14 patients, 5 patients were classical PKU, 3 were atypical PKU, and 6 were mild PKU. All of these patients had at least one mutation in the regulatory domain. Patients were given 10 mg/kg BH4, and the response of blood phenylalanine (Phe) levels was monitored following treatment. The level of blood Phe decreased after BH4 treatment in all of the patients. These studies suggest that mutations in the regulatory domain also responded to BH4 even if the patient had classical PKU.


Asunto(s)
Biopterinas/análogos & derivados , Mutación , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/genética , Adolescente , Adulto , Sustitución de Aminoácidos , Biopterinas/uso terapéutico , Niño , Análisis Mutacional de ADN , Humanos , Persona de Mediana Edad , Modelos Moleculares , Fenilalanina Hidroxilasa/química , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/enzimología , Conformación Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Eliminación de Secuencia
13.
Exp Anim ; 56(2): 161-5, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17460362

RESUMEN

The Spontaneously Epileptic Rat (SER), a double-mutant for tremor and zitter mutations, shows spontaneous occurrences of absence-like and tonic seizures. Several lines of evidence suggest that the combined effect of Aspa and Atrn mutations is the most likely cause of the epileptic phenotype of the SER. To address this issue, we produced a new double-mutant mouse line carrying both homozygous Aspa-knockout and Atrn(mg-3J) mutant alleles. The Aspa/Atrn double-mutant mice exhibited absence-like and tonic seizures that were characterized by the appearance of 5-7 Hz spike-wave-like complexes and low voltage fast waves on EEGs. These results demonstrate directly that the simultaneous loss of the Aspa and Atrn gene functions causes epileptic seizures in the mouse and suggest that both Aspa and Atrn deficiencies might be responsible for epileptic seizures in the SER.


Asunto(s)
Amidohidrolasas/deficiencia , Epilepsia Tipo Ausencia/metabolismo , Proteínas de la Membrana/deficiencia , Convulsiones/metabolismo , Amidohidrolasas/genética , Animales , Electroencefalografía , Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/fisiopatología , Femenino , Silenciador del Gen , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Mutantes , Convulsiones/genética , Convulsiones/fisiopatología
14.
JCI Insight ; 2(3): e90807, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28194442

RESUMEN

Canavan disease (CD) is a debilitating and lethal leukodystrophy caused by mutations in the aspartoacylase (ASPA) gene and the resulting defect in N-acetylaspartate (NAA) metabolism in the CNS and peripheral tissues. Recombinant adeno-associated virus (rAAV) has the ability to cross the blood-brain barrier and widely transduce the CNS. We developed a rAAV-based and optimized gene replacement therapy, which achieves early, complete, and sustained rescue of the lethal disease phenotype in CD mice. Our treatment results in a super-mouse phenotype, increasing motor performance of treated CD mice beyond that of WT control mice. We demonstrate that this rescue is oligodendrocyte independent, and that gene correction in astrocytes is sufficient, suggesting that the establishment of an astrocyte-based alternative metabolic sink for NAA is a key mechanism for efficacious disease rescue and the super-mouse phenotype. Importantly, the use of clinically translatable high-field imaging tools enables the noninvasive monitoring and prediction of therapeutic outcomes for CD and might enable further investigation of NAA-related cognitive function.


Asunto(s)
Amidohidrolasas/genética , Enfermedad de Canavan/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Amidohidrolasas/metabolismo , Animales , Ácido Aspártico/análogos & derivados , Barrera Hematoencefálica/metabolismo , Enfermedad de Canavan/genética , Enfermedad de Canavan/metabolismo , Sistema Nervioso Central , Dependovirus/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Oligodendroglía/metabolismo
15.
Genome Med ; 9(1): 83, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28934986

RESUMEN

BACKGROUND: Exon-targeted microarrays can detect small (<1000 bp) intragenic copy number variants (CNVs), including those that affect only a single exon. This genome-wide high-sensitivity approach increases the molecular diagnosis for conditions with known disease-associated genes, enables better genotype-phenotype correlations, and facilitates variant allele detection allowing novel disease gene discovery. METHODS: We retrospectively analyzed data from 63,127 patients referred for clinical chromosomal microarray analysis (CMA) at Baylor Genetics laboratories, including 46,755 individuals tested using exon-targeted arrays, from 2007 to 2017. Small CNVs harboring a single gene or two to five non-disease-associated genes were identified; the genes involved were evaluated for a potential disease association. RESULTS: In this clinical population, among rare CNVs involving any single gene reported in 7200 patients (11%), we identified 145 de novo autosomal CNVs (117 losses and 28 intragenic gains), 257 X-linked deletion CNVs in males, and 1049 inherited autosomal CNVs (878 losses and 171 intragenic gains); 111 known disease genes were potentially disrupted by de novo autosomal or X-linked (in males) single-gene CNVs. Ninety-one genes, either recently proposed as candidate disease genes or not yet associated with diseases, were disrupted by 147 single-gene CNVs, including 37 de novo deletions and ten de novo intragenic duplications on autosomes and 100 X-linked CNVs in males. Clinical features in individuals with de novo or X-linked CNVs encompassing at most five genes (224 bp to 1.6 Mb in size) were compared to those in individuals with larger-sized deletions (up to 5 Mb in size) in the internal CMA database or loss-of-function single nucleotide variants (SNVs) detected by clinical or research whole-exome sequencing (WES). This enabled the identification of recently published genes (BPTF, NONO, PSMD12, TANGO2, and TRIP12), novel candidate disease genes (ARGLU1 and STK3), and further confirmation of disease association for two recently proposed disease genes (MEIS2 and PTCHD1). Notably, exon-targeted CMA detected several pathogenic single-exon CNVs missed by clinical WES analyses. CONCLUSIONS: Together, these data document the efficacy of exon-targeted CMA for detection of genic and exonic CNVs, complementing and extending WES in clinical diagnostics, and the potential for discovery of novel disease genes by genome-wide assay.


Asunto(s)
Variaciones en el Número de Copia de ADN , Exones , Enfermedades Genéticas Congénitas , Estudios de Cohortes , Genoma Humano , Proteínas de Homeodominio/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Trastornos del Neurodesarrollo/genética , Proteínas Serina-Treonina Quinasas/genética , Estudios Retrospectivos , Serina-Treonina Quinasa 3 , Factores de Transcripción/genética , Secuenciación Completa del Genoma
16.
Adv Exp Med Biol ; 576: 77-93; discussion 361-3, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16802706

RESUMEN

Canavan disease (CD) is an autosomal recessive disorder, characterized by spongy degeneration of the brain. Patients with CD have aspartoacylase (ASPA) deficiency, which results accumulation of N-acetylaspartic acid (NAA) in the brain and elevated excretion of urinary NAA. Clinically, patients with CD have macrocephaly, mental retardation and hypotonia. A knockout mouse for CD which was engineered, also has ASPA deficiency and elevated NAA. Molecular studies of the mouse brain showed abnormal expression of multiple genes in addition to ASPA deficiency. Adenoassociated virus mediated gene transfer and stem cell therapy in the knockout mouse are the latest attempts to alter pathophysiology in the CD mouse.


Asunto(s)
Amidohidrolasas , Enfermedad de Canavan , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Enfermedad de Canavan/genética , Enfermedad de Canavan/metabolismo , Enfermedad de Canavan/patología , Enfermedad de Canavan/terapia , Dipéptidos/metabolismo , Marcación de Gen , Terapia Genética , Ácido Glutámico/metabolismo , Humanos , Ratones , Ratones Noqueados , Fenotipo , Trasplante de Células Madre , Ácido gamma-Aminobutírico/metabolismo
17.
Eur J Med Genet ; 59(9): 470-3, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27238888

RESUMEN

We report an 8-year-old female with autism spectrum disorder (ASD), intellectual disability and speech delay who was found to carry a de novo 82 kb deletion of chromosome Xq11.1-11.2 involving the ARHGEF9 gene on chromosomal microarray. So far, 11 patients with point mutations, disruptions due to chromosomal rearrangements and deletions involving ARHGEF9 have been reported in the literature. ARHGEF9-related disorders comprise a wide phenotypic spectrum, including behavior disorders, autism spectrum disorder, intellectual disability, hyperekplexia and infantile epileptic encephalopathy. ARHGEF9 encodes for collybistin which plays an important role in post synaptic clustering of glycine and inhibitory gamma-aminobutyric acid receptors along with its scaffolding partner, gephyrin. The reduction of inhibitory receptor clusters in brain has been proposed as a plausible underlying pathophysiological mechanism. With this report, we provide further evidence for the role of ARHGEF9 in neurocognitive function, its implication in ASD, and review the clinical features of previously published individuals with ARHGEF9-related intellectual disability.


Asunto(s)
Trastorno del Espectro Autista/genética , Cromosomas Humanos X , Factores de Intercambio de Guanina Nucleótido Rho/genética , Niño , Deleción Cromosómica , Femenino , Humanos , Recién Nacido , Embarazo
18.
Pediatrics ; 137(4)2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27020793

RESUMEN

BACKGROUND: Viral upper and lower respiratory tract infections (URI, LRI) are common in infants. We determined the prevalence of viral URI and its complications, including acute otitis media (AOM) and LRI, and assessed the effect of bacterial-viral interactions, and genetic and environmental risks on AOM development. METHODS: Healthy infants were enrolled from near birth and followed to the first episode of AOM up to 12 months of age. Nasopharyngeal specimens were collected at monthly intervals (months 1-6, 9) and during viral URI episodes for bacterial culture and viral polymerase chain reaction studies. Subjects were followed closely for AOM development. RESULTS: A total of 367 infants were followed for 286 child-years; 887 URI (305 infants) and 180 AOM episodes (143 infants) were documented. Prevalence of URI, LRI, and AOM in the first year was 3.2, 0.25, and 0.67 per child-year, respectively. Cumulative AOM incidence by ages 3, 6, and 12 months was 6%, 23%, and 46%. Infants with and without AOM had 4.7 and 2.3 URI episodes per child-year, respectively (P < .002). Pathogenic bacterial colonization rates by month were significantly higher in infants with AOM (P < .005). Breastfeeding reduced both URI and AOM risks (P < .05). Significant bacterial-viral interactions occurred with Moraxella catarrhalis and a variety of respiratory viruses and altered URI and AOM risks. CONCLUSIONS: Almost half of infants experienced AOM by age 1. Important AOM risk factors included frequent viral URI, pathogenic bacterial colonization, and lack of breastfeeding. Bacterial-viral interactions may play a significant role in AOM pathogenesis and deserve further investigation.


Asunto(s)
Otitis Media/etiología , Infecciones del Sistema Respiratorio/complicaciones , Virosis/complicaciones , Enfermedad Aguda , Infecciones Bacterianas/complicaciones , Comorbilidad , Femenino , Humanos , Incidencia , Lactante , Estudios Longitudinales , Masculino , Nasofaringe/microbiología , Otitis Media/epidemiología , Factores de Riesgo
19.
Neurochem Int ; 46(8): 595-9, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15863237

RESUMEN

Phenylketonuria (PKU) is an inborn error of amino acid metabolism. Phenylalanine hydroxylase (PAH) deficiency results in accumulation of phenylalanine (Phe) in the brain and leads to pathophysiological abnormalities including cognitive defect, if Phe diet is not restricted. Neuronatin and 4-nitrophenylphosphatase domain and non-neuronal SNAP25-like protein homolog 1 (NIPSNAP1) reportedly have role in memory. Therefore, gene expression was examined in the brain of mouse model for PKU. Microarray expression analysis revealed reduced expression of calpastatin, NIPSNAP 1, rabaptin-5 and minopontin genes and overexpression of neuronatin gene in the PKU mouse brain. Altered expression of these genes was further confirmed by one-step real time RT-PCR analysis. Western blot analysis of the mouse brain showed reduced levels of calpastatin and rabaptin-5 and higher amount of neuronatin in PKU compared to the wild type. These observations in the PKU mouse brain suggest that altered expression of these genes resulting in abnormal proteome. These changes in the PKU mouse brain are likely to contribute cognitive impairment seen in the PKU mouse, if documented also in patients with PKU.


Asunto(s)
Encéfalo/metabolismo , Trastornos del Conocimiento/metabolismo , Perfilación de la Expresión Génica , Expresión Génica/genética , Proteínas del Tejido Nervioso/metabolismo , Fenilcetonurias/metabolismo , Animales , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/genética , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteopontina , Fenilcetonurias/complicaciones , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/metabolismo , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Regulación hacia Arriba/fisiología , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
20.
Neurosci Lett ; 382(3): 323-6, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15925112

RESUMEN

Phenylketonuria (PKU) is an inborn error of amino acid metabolism. Phenylalanine hydroxylase (PAH) mutations resulting reduced enzyme levels lead to accumulation of phenylalanine (Phe) in brain, if Phe diet is not restricted. Patients with PKU show neurophysiological abnormalities including demyelination and cognitive defect. How PAH defect causes events seen in PKU is not obvious. Therefore, expression analysis was performed in the brain of a mouse model for PKU. Microarray expression profile of the brain showed lower expression of myocilin (Myoc) in the PKU mouse. Reduced expression of Myoc was further confirmed by one-step real-time RT-PCR. Western blotting analysis of the brain using equal quantities of protein showed a thin band in PKU compared to a prominent band in the wild type brain. In addition, expression of genes associated with transcription was found to be altered in the PKU mouse brain as observed by microarray analysis. These data suggest that PAH defect alters other genes expression likely to contribute neurophysiological abnormalities seen in the mouse, if documented also in patients with PKU.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Proteínas del Ojo/biosíntesis , Glicoproteínas/biosíntesis , Fenilcetonurias/fisiopatología , Animales , Western Blotting , Modelos Animales de Enfermedad , Expresión Génica , Regulación de la Expresión Génica , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/metabolismo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA