Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 23(1): 123-132, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28115742

RESUMO

Large-scale consortia mapping the genomic risk architectures of schizophrenia provide vast amounts of molecular information, with largely unexplored therapeutic potential. We harnessed publically available information from the Psychiatric Genomics Consortium, and report myocyte enhancer factor 2C (MEF2C) motif enrichment in sequences surrounding the top scoring single-nucleotide polymorphisms within risk loci contributing by individual small effect to disease heritability. Chromatin profiling at base-pair resolution in neuronal nucleosomes extracted from prefrontal cortex of 34 subjects, including 17 cases diagnosed with schizophrenia, revealed MEF2C motif enrichment within cis-regulatory sequences, including neuron-specific promoters and superenhancers, affected by histone H3K4 hypermethylation in disease cases. Vector-induced short- and long-term Mef2c upregulation in mouse prefrontal projection neurons consistently resulted in enhanced cognitive performance in working memory and object recognition paradigms at baseline and after psychotogenic drug challenge, in conjunction with remodeling of local connectivity. Neuronal genome tagging in vivo by Mef2c-Dam adenine methyltransferase fusion protein confirmed the link between cognitive enhancement and MEF2C occupancy at promoters harboring canonical and variant MEF2C motifs. The multilayered integrative approaches presented here provide a roadmap to uncover the therapeutic potential of transcriptional regulators for schizophrenia and related disorders.


Assuntos
Transtornos Cognitivos , Regulação da Expressão Gênica/genética , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/complicações , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Imunoprecipitação da Cromatina , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/terapia , Biologia Computacional , Modelos Animais de Doenças , Epigenômica/métodos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Esquizofrenia/genética , Esquizofrenia/patologia , Transdução Genética
2.
Mol Psychiatry ; 23(2): 271-281, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-27752082

RESUMO

The brain-specific tyrosine phosphatase, STEP (STriatal-Enriched protein tyrosine Phosphatase) is an important regulator of synaptic function. STEP normally opposes synaptic strengthening by increasing N-methyl D-aspartate glutamate receptor (NMDAR) internalization through dephosphorylation of GluN2B and inactivation of the kinases extracellular signal-regulated kinase 1/2 and Fyn. Here we show that STEP61 is elevated in the cortex in the Nrg1+/- knockout mouse model of schizophrenia (SZ). Genetic reduction or pharmacological inhibition of STEP prevents the loss of NMDARs from synaptic membranes and reverses behavioral deficits in Nrg1+/- mice. STEP61 protein is also increased in cortical lysates from the central nervous system-specific ErbB2/4 mouse model of SZ, as well as in human induced pluripotent stem cell (hiPSC)-derived forebrain neurons and Ngn2-induced excitatory neurons, from two independent SZ patient cohorts. In these selected SZ models, increased STEP61 protein levels likely reflect reduced ubiquitination and degradation. These convergent findings from mouse and hiPSC SZ models provide evidence for STEP61 dysfunction in SZ.


Assuntos
Proteínas Tirosina Fosfatases/fisiologia , Esquizofrenia/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuregulina-1/genética , Neurônios/metabolismo , Fosforilação , Proteínas Tirosina Fosfatases/genética , Ratos , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/genética , Ubiquitinação
3.
Mol Cell Neurosci ; 82: 157-166, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28549865

RESUMO

CRISPR/Cas9 technology has transformed our ability to manipulate the genome and epigenome, from efficient genomic editing to targeted localization of effectors to specific loci. Through the manipulation of DNA- and histone-modifying enzyme activities, activation or repression of gene expression, and targeting of transcriptional regulators, the role of gene-regulatory and epigenetic pathways in basic biology and disease processes can be directly queried. Here, we discuss emerging CRISPR-based methodologies, with specific consideration of neurobiological applications of human induced pluripotent stem cell (hiPSC)-based models.


Assuntos
Encéfalo/crescimento & desenvolvimento , Sistemas CRISPR-Cas/genética , Edição de Genes , Expressão Gênica/genética , Células-Tronco Pluripotentes Induzidas/citologia , Encefalopatias/terapia , Edição de Genes/métodos , Humanos
4.
Schizophr Res ; 185: 17-32, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28094170

RESUMO

The watershed-hypothesis of schizophrenia asserts that over 200 different mutations dysregulate distinct pathways that converge on an unspecified common mechanism(s) that controls disease ontogeny. Consistent with this hypothesis, our RNA-sequencing of neuron committed cells (NCCs) differentiated from established iPSCs of 4 schizophrenia patients and 4 control subjects uncovered a dysregulated transcriptome of 1349 mRNAs common to all patients. Data reveals a global dysregulation of developmental genome, deconstruction of coordinated mRNA networks, and the formation of aberrant, new coordinated mRNA networks indicating a concerted action of the responsible factor(s). Sequencing of miRNA transcriptomes demonstrated an overexpression of 16 miRNAs and deconstruction of interactive miRNA-mRNA networks in schizophrenia NCCs. ChiPseq revealed that the nuclear (n) form of FGFR1, a pan-ontogenic regulator, is overexpressed in schizophrenia NCCs and overtargets dysregulated mRNA and miRNA genes. The nFGFR1 targeted 54% of all human gene promoters and 84.4% of schizophrenia dysregulated genes. The upregulated genes reside within major developmental pathways that control neurogenesis and neuron formation, whereas downregulated genes are involved in oligodendrogenesis. Our results indicate (i) an early (preneuronal) genomic etiology of schizophrenia, (ii) dysregulated genes and new coordinated gene networks are common to unrelated cases of schizophrenia, (iii) gene dysregulations are accompanied by increased nFGFR1-genome interactions, and (iv) modeling of increased nFGFR1 by an overexpression of a nFGFR1 lead to up or downregulation of selected genes as observed in schizophrenia NCCs. Together our results designate nFGFR1 signaling as a potential common dysregulated mechanism in investigated patients and potential therapeutic target in schizophrenia.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Genoma/genética , Mutação/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Esquizofrenia/genética , Transdução de Sinais/genética , Adulto , Diferenciação Celular , Células Cultivadas , Feminino , Redes Reguladoras de Genes , Genômica , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Receptor Notch1/metabolismo , Esquizofrenia/patologia , Transcriptoma , Adulto Jovem
5.
Transl Psychiatry ; 7(11): 6, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-30446636

RESUMO

Studies of induced pluripotent stem cells (iPSCs) from schizophrenia patients and control individuals revealed that the disorder is programmed at the preneuronal stage, involves a common dysregulated mRNA transcriptome, and identified Integrative Nuclear FGFR1 Signaling a common dysregulated mechanism. We used human embryonic stem cell (hESC) and iPSC-derived cerebral organoids from four controls and three schizophrenia patients to model the first trimester of in utero brain development. The schizophrenia organoids revealed an abnormal scattering of proliferating Ki67+ neural progenitor cells (NPCs) from the ventricular zone (VZ), throughout the intermediate (IZ) and cortical (CZ) zones. TBR1 pioneer neurons and reelin, which guides cortico-petal migration, were restricted from the schizophrenia cortex. The maturing neurons were abundantly developed in the subcortical regions, but were depleted from the schizophrenia cortex. The decreased intracortical connectivity was denoted by changes in the orientation and morphology of calretinin interneurons. In schizophrenia organoids, nuclear (n)FGFR1 was abundantly expressed by developing subcortical cells, but was depleted from the neuronal committed cells (NCCs) of the CZ. Transfection of dominant negative and constitutively active nFGFR1 caused widespread disruption of the neuro-ontogenic gene networks in hESC-derived NPCs and NCCs. The fgfr1 gene was the most prominent FGFR gene expressed in NPCs and NCCs, and blocking with PD173074 reproduced both the loss of nFGFR1 and cortical neuronal maturation in hESC cerebral organoids. We report for the first time, progression of the cortical malformation in schizophrenia and link it to altered FGFR1 signaling. Targeting INFS may offer a preventive treatment of schizophrenia.


Assuntos
Córtex Cerebral/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Esquizofrenia/patologia , Calbindina 2/metabolismo , Córtex Cerebral/metabolismo , Células-Tronco Embrionárias/patologia , Humanos , Interneurônios/metabolismo , Interneurônios/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Proteína Reelina , Esquizofrenia/genética , Esquizofrenia/metabolismo
6.
Transl Psychiatry ; 5: e662, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26485546

RESUMO

The genetic and epigenetic factors contributing to risk for schizophrenia (SZ) remain unresolved. Here we demonstrate, for the first time, perturbed global protein translation in human-induced pluripotent stem cell (hiPSC)-derived forebrain neural progenitor cells (NPCs) from four SZ patients relative to six unaffected controls. We report increased total protein levels and protein synthesis, together with two independent sets of quantitative mass spectrometry evidence indicating markedly increased levels of ribosomal and translation initiation and elongation factor proteins, in SZ hiPSC NPCs. We posit that perturbed levels of global protein synthesis in SZ hiPSC NPCs represent a novel post-transcriptional mechanism that might contribute to disease progression.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Prosencéfalo/metabolismo , Esquizofrenia/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos
8.
Mol Psychiatry ; 20(3): 361-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24686136

RESUMO

Consistent with recent reports indicating that neurons differentiated in vitro from human-induced pluripotent stem cells (hiPSCs) are immature relative to those in the human brain, gene expression comparisons of our hiPSC-derived neurons to the Allen BrainSpan Atlas indicate that they most resemble fetal brain tissue. This finding suggests that, rather than modeling the late features of schizophrenia (SZ), hiPSC-based models may be better suited for the study of disease predisposition. We now report that a significant fraction of the gene signature of SZ hiPSC-derived neurons is conserved in SZ hiPSC neural progenitor cells (NPCs). We used two independent discovery-based approaches-microarray gene expression and stable isotope labeling by amino acids in cell culture (SILAC) quantitative proteomic mass spectrometry analyses-to identify cellular phenotypes in SZ hiPSC NPCs from four SZ patients. From our findings that SZ hiPSC NPCs show abnormal gene expression and protein levels related to cytoskeletal remodeling and oxidative stress, we predicted, and subsequently observed, aberrant migration and increased oxidative stress in SZ hiPSC NPCs. These reproducible NPC phenotypes were identified through scalable assays that can be applied to expanded cohorts of SZ patients, making them a potentially valuable tool with which to study the developmental mechanisms contributing to SZ.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Neurais/metabolismo , Células-Tronco Pluripotentes/fisiologia , Prosencéfalo/patologia , Esquizofrenia/patologia , Adulto , Animais , Antipsicóticos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular , Células Cultivadas , Feminino , Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Fenótipo , Células-Tronco Pluripotentes/efeitos dos fármacos , Proteômica , Espécies Reativas de Oxigênio/metabolismo , Adulto Jovem
9.
Mol Psychiatry ; 17(12): 1239-53, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22472874

RESUMO

Although psychiatric disorders such as autism spectrum disorders, schizophrenia and bipolar disorder affect a number of brain regions and produce a complex array of clinical symptoms, basic phenotypes likely exist at the level of single neurons and simple networks. Being highly heritable, it is hypothesized that these disorders are amenable to cell-based studies in vitro. Using induced pluripotent stem cell-derived neurons and/or induced neurons from fibroblasts, limitless numbers of live human neurons can now be generated from patients with a genetic background permissive to the disease state. We predict that cell-based studies will ultimately contribute to our understanding of the initiation, progression and treatment of these psychiatric disorders.


Assuntos
Fibroblastos/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Transtornos Mentais/fisiopatologia , Vias Neurais/fisiopatologia , Animais , Fibroblastos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Neurológicos , Neurônios/patologia , Neurônios/fisiologia , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...