Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Am J Hum Genet ; 95(5): 490-508, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25307298

RESUMEN

Neurodevelopmental disorders (NDDs) are caused by mutations in diverse genes involved in different cellular functions, although there can be crosstalk, or convergence, between molecular pathways affected by different NDDs. To assess molecular convergence, we generated human neural progenitor cell models of 9q34 deletion syndrome, caused by haploinsufficiency of EHMT1, and 18q21 deletion syndrome, caused by haploinsufficiency of TCF4. Using next-generation RNA sequencing, methylation sequencing, chromatin immunoprecipitation sequencing, and whole-genome miRNA analysis, we identified several levels of convergence. We found mRNA and miRNA expression patterns that were more characteristic of differentiating cells than of proliferating cells, and we identified CpG clusters that had similar methylation states in both models of reduced gene dosage. There was significant overlap of gene targets of TCF4 and EHMT1, whereby 8.3% of TCF4 gene targets and 4.2% of EHMT1 gene targets were identical. These data suggest that 18q21 and 9q34 deletion syndromes show significant molecular convergence but distinct expression and methylation profiles. Common intersection points might highlight the most salient features of disease and provide avenues for similar treatments for NDDs caused by different genetic mutations.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Trastornos de los Cromosomas/genética , Anomalías Craneofaciales/genética , Evolución Molecular , Haploinsuficiencia/genética , Cardiopatías Congénitas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Células-Madre Neurales , Factores de Transcripción/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Deleción Cromosómica , Cromosomas Humanos Par 18/genética , Cromosomas Humanos Par 9/genética , Metilación de ADN , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , MicroARNs/genética , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN , Factor de Transcripción 4
2.
Am J Med Genet A ; 170A(5): 1225-35, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26789910

RESUMEN

Mutations in chromodomain helicase DNA-binding domain 8 (CHD8) have been identified in independent genotyping studies of autism spectrum disorder. To better understand the phenotype associated with CHD8 mutations, we genotyped all CHD8 exons in carefully assessed cohorts of autism (n = 142), schizophrenia (SCZ; n = 143), and intellectual disability (ID; n = 94). We identified one frameshift mutation, seven non-synonymous variants, and six synonymous variants. The frameshift mutation, p.Asn2092Lysfs*2, which creates a premature stop codon leading to the loss of 212 amino acids of the protein, was from an autism case on whom we present multiple clinical assessments and pharmacological treatments spanning more than 10 years. RNA and protein analysis support a model where the transcript generated from the mutant allele results in haploinsufficiency of CHD8. This case report supports the association of CHD8 mutations with classical autism, macrocephaly, infantile hypotonia, speech delay, lack of major ID, and psychopathology in late adolescence caused by insufficient dosage of CHD8. Review of 16 other CHD8 mutation cases suggests that clinical features and their severity vary considerably across individuals; however, these data support a CHD8 mutation syndrome, further highlighting the importance of genomic medicine to guide clinical assessment and treatment.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas de Unión al ADN/genética , Esquizofrenia/genética , Factores de Transcripción/genética , Adolescente , Trastorno del Espectro Autista/fisiopatología , Niño , Preescolar , Hibridación Genómica Comparativa , Exones/genética , Femenino , Mutación del Sistema de Lectura , Regulación del Desarrollo de la Expresión Génica , Genotipo , Humanos , Masculino , Esquizofrenia/fisiopatología
3.
Hum Genet ; 134(10): 1037-53, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26194112

RESUMEN

Several neurodevelopmental disorders (NDDs) are caused by mutations in genes expressed in fetal brain, but little is known about these same genes in adult human brain. Here, we test the hypothesis that genes associated with NDDs continue to have a role in adult human brain to explore the idea that NDD symptoms may be partially a result of their adult function rather than just their neurodevelopmental function. To demonstrate adult brain function, we performed expression analyses and ChIPseq in human neural stem cell(NSC) lines at different developmental stages and adult human brain, targeting two genes associated with NDDs, SATB2 and EHMT1, and the WNT signaling gene TCF7L2, which has not been associated with NDDs. Analysis of DNA interaction sites in neural stem cells reveals high (40-50 %) overlap between proliferating and differentiating cells for each gene in temporal space. Studies in adult brain demonstrate that consensus sites are similar to NSCs but occur at different genomic locations. We also performed expression analyses using BrainSpan data for NDD-associated genes SATB2, EHMT1, FMR1, MECP2, MBD5, CTNND2, RAI1, CHD8, GRIN2A, GRIN2B, TCF4, SCN2A, and DYRK1A and find high expression of these genes in adult brain, at least comparable to developing human brain, confirming that genes associated with NDDs likely have a role in adult tissue. Adult function of genes associated with NDDs might be important in clinical disease presentation and may be suitable targets for therapeutic intervention.


Asunto(s)
Trastornos del Neurodesarrollo/genética , Adulto , Secuencia de Bases , Línea Celular , Secuencia de Consenso , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Persona de Mediana Edad , Células-Madre Neurales/fisiología , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Adulto Joven
4.
Am J Hum Genet ; 91(6): 1128-34, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23217328

RESUMEN

Large intergenic noncoding (linc) RNAs represent a newly described class of ribonucleic acid whose importance in human disease remains undefined. We identified a severely developmentally delayed 16-year-old female with karyotype 46,XX,t(2;11)(p25.1;p15.1)dn in the absence of clinically significant copy number variants (CNVs). DNA capture followed by next-generation sequencing of the translocation breakpoints revealed disruption of a single noncoding gene on chromosome 2, LINC00299, whose RNA product is expressed in all tissues measured, but most abundantly in brain. Among a series of additional, unrelated subjects referred for clinical diagnostic testing who showed CNV affecting this locus, we identified four with exon-crossing deletions in association with neurodevelopmental abnormalities. No disruption of the LINC00299 coding sequence was seen in almost 14,000 control subjects. Together, these subjects with disruption of LINC00299 implicate this particular noncoding RNA in brain development and raise the possibility that, as a class, abnormalities of lincRNAs may play a significant role in human developmental disorders.


Asunto(s)
Discapacidades del Desarrollo/genética , Mutación , ARN Largo no Codificante/genética , Adolescente , Empalme Alternativo , Secuencia de Bases , Puntos de Rotura del Cromosoma , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 2 , Femenino , Orden Génico , Humanos , Linfocitos/metabolismo , Datos de Secuencia Molecular , Células-Madre Neurales/metabolismo , Translocación Genética
5.
Stem Cell Reports ; 16(7): 1749-1762, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34214487

RESUMEN

Mutations in HPRT1, a gene encoding a rate-limiting enzyme for purine salvage, cause Lesch-Nyhan disease which is characterized by self-injury and motor impairments. We leveraged stem cell and genetic engineering technologies to model the disease in isogenic and patient-derived forebrain and midbrain cell types. Dopaminergic progenitor cells deficient in HPRT showed decreased intensity of all developmental cell-fate markers measured. Metabolic analyses revealed significant loss of all purine derivatives, except hypoxanthine, and impaired glycolysis and oxidative phosphorylation. real-time glucose tracing demonstrated increased shunting to the pentose phosphate pathway for de novo purine synthesis at the expense of ATP production. Purine depletion in dopaminergic progenitor cells resulted in loss of RHEB, impairing mTORC1 activation. These data demonstrate dopaminergic-specific effects of purine salvage deficiency and unexpectedly reveal that dopaminergic progenitor cells are programmed to a high-energy state prior to higher energy demands of terminally differentiated cells.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Metabolismo Energético , Síndrome de Lesch-Nyhan/metabolismo , Síndrome de Lesch-Nyhan/patología , Mesencéfalo/patología , Biomarcadores/metabolismo , Linaje de la Célula , Corteza Cerebral/patología , Glucosa/metabolismo , Glucólisis , Humanos , Hipoxantina Fosforribosiltransferasa/deficiencia , Síndrome de Lesch-Nyhan/enzimología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células-Madre Neurales/metabolismo , Fosforilación Oxidativa , Vía de Pentosa Fosfato , Purinas/metabolismo
6.
Stem Cells Transl Med ; 6(3): 886-896, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28170165

RESUMEN

The development of targeted therapeutics for rare neurodevelopmental disorders (NDDs) faces significant challenges due to the scarcity of subjects and the difficulty of obtaining human neural cells. Here, we illustrate a rapid, simple protocol by which patient derived cells can be reprogrammed to induced pluripotent stem cells (iPSCs) using an episomal vector and differentiated into neurons. Using this platform enables patient somatic cells to be converted to physiologically active neurons in less than two months with minimal labor. This platform includes a method to combine somatic cell reprogramming with CRISPR/Cas9 gene editing at single cell resolution, which enables the concurrent development of clonal knockout or knock-in models that can be used as isogenic control lines. This platform reduces the logistical barrier for using iPSC technology, allows for the development of appropriate control lines for use in rare neurodevelopmental disease research, and establishes a fundamental component to targeted therapeutics and precision medicine. Stem Cells Translational Medicine 2017;6:886-896.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Edición Génica , Modelos Biológicos , Trastornos del Neurodesarrollo/patología , Secuencia de Bases , Diferenciación Celular , Fibroblastos/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mesencéfalo/patología , Neuronas/patología , Prosencéfalo/patología
7.
Mol Syndromol ; 7(6): 302-311, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27920633

RESUMEN

Lesch-Nyhan syndrome (LNS) is a rare X-linked disorder caused by mutations in HPRT1, an important enzyme in the purine salvage pathway. Symptoms of LNS include dystonia, gout, intellectual disability, and self-mutilation. Despite having been characterized over 50 years ago, it remains unclear precisely how deficits in hypoxanthine and guanine recycling can lead to such a profound neurological phenotype. Several studies have proposed different hypotheses regarding the etiology of this disease, and several treatments have been tried in patients, though none have led to a satisfactory explanation of the disease. New technologies such as next-generation sequencing, optogenetics, genome editing, and induced pluripotent stem cells provide a unique opportunity to map the precise sequential pathways leading from genotype to phenotype.

8.
Pediatr Clin North Am ; 62(3): 619-31, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26022166

RESUMEN

Self-injury is a complex and poorly understood behavior observed in people with psychopathology or neurodevelopmental disorders (NDD). Despite the differences in etiology and progression of these distinct disease domains, it is possible that overlapping molecular pathways underlie the expression of self-injurious behaviors (SIBs). This review outlines the similarities and differences at the behavioural and molecular level, where SIBs in both conditions may involve opioid, nucleoside, and dopamine signalling. These points of convergence have important implications for treatment and research of SIB in both populations.


Asunto(s)
Discapacidades del Desarrollo/psicología , Trastornos Mentales/psicología , Conducta Autodestructiva/psicología , Niño , Discapacidades del Desarrollo/epidemiología , Humanos , Trastornos Mentales/epidemiología , Prevalencia , Factores de Riesgo , Conducta Autodestructiva/epidemiología
9.
Nat Med ; 20(7): 764-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24908571

RESUMEN

Major depressive disorder (MDD) is a prevalent mood disorder that is associated with differential prefrontal brain expression patterns. Treatment of MDD includes a variety of biopsychosocial approaches. In medical practice, antidepressant drugs are the most common treatment for depressive episodes, and they are among the most prescribed medications in North America. Although antidepressants are clearly effective, particularly for moderate to severe depressive episodes, there is variability in how individuals respond to antidepressant treatment. Failure to respond has individual, economic and social consequences for patients and their families. Several lines of evidence demonstrate that genes are regulated through the activity of microRNAs (miRNAs), which act as fine-tuners and on-off switches of gene expression. Here we report on complementary studies using postmortem human brain samples, cellular assays and samples from clinical trials of patients with depression and show that miR-1202, a miRNA specific to primates and enriched in the human brain, is differentially expressed in individuals with depression. Additionally, miR-1202 regulates expression of the gene encoding metabotropic glutamate receptor-4 (GRM4) and predicts antidepressant response at baseline. These results suggest that miR-1202 is associated with the pathophysiology of depression and is a potential target for new antidepressant treatments.


Asunto(s)
Antidepresivos/uso terapéutico , Encéfalo/metabolismo , Trastorno Depresivo Mayor/genética , MicroARNs/fisiología , Línea Celular , Trastorno Depresivo Mayor/tratamiento farmacológico , Humanos , MicroARNs/metabolismo
10.
Arch Gen Psychiatry ; 69(12): 1238-46, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23044507

RESUMEN

CONTEXT Brain-derived neurotrophic factor (BDNF) is suspected of being a causative factor in psychiatric disorders based on case reports or studies involving large structural anomalies. OBJECTIVE To determine the involvement of BDNF in human psychopathology. DESIGN Case-control study. SETTING Microarray-based comparative genomic hybridization data from 7 molecular diagnostic centers including 38 550 affected subjects and 28 705 unaffected subjects. PATIENTS Subjects referred to diagnostic screening centers for microarray-based comparative genomic hybridization for physical or cognitive impairment. MAIN OUTCOME MEASURES Genomic copy number gains and losses. RESULTS We report 5 individuals with psychopathology and genomic deletion of a critical region including BDNF. The defined critical region was never disrupted in control subjects or diagnostic cases without developmental abnormalities. CONCLUSION Hemizygosity of the BDNF region contributes to variable psychiatric phenotypes including anxiety, behavioral, and mood disorders.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA