Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biol Psychiatry ; 95(2): 187-198, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-37454787

RESUMEN

BACKGROUND: Converging evidence from large-scale genetic and postmortem studies highlights the role of aberrant neurotransmission and genetic regulation in brain-related disorders. However, identifying neuronal activity-regulated transcriptional programs in the human brain and understanding how changes contribute to disease remain challenging. METHODS: To better understand how the activity-dependent regulome contributes to risk for brain-related disorders, we profiled the transcriptomic and epigenomic changes following neuronal depolarization in human induced pluripotent stem cell-derived glutamatergic neurons (NGN2) from 6 patients with schizophrenia and 5 control participants. RESULTS: Multiomic data integration associated global patterns of chromatin accessibility with gene expression and identified enhancer-promoter interactions in glutamatergic neurons. Within 1 hour of potassium chloride-induced depolarization, independent of diagnosis, glutamatergic neurons displayed substantial activity-dependent changes in the expression of genes regulating synaptic function. Depolarization-induced changes in the regulome revealed significant heritability enrichment for schizophrenia and Parkinson's disease, adding to mounting evidence that sequence variation within activation-dependent regulatory elements contributes to the genetic risk for brain-related disorders. Gene coexpression network analysis elucidated interactions among activity-dependent and disease-associated genes and pointed to a key driver (NAV3) that interacted with multiple genes involved in axon guidance. CONCLUSIONS: Overall, we demonstrated that deciphering the activity-dependent regulome in glutamatergic neurons reveals novel targets for advanced diagnosis and therapy.


Asunto(s)
Células Madre Pluripotentes Inducidas , Esquizofrenia , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Encéfalo
2.
Sci Transl Med ; 15(693): eadf1147, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37099632

RESUMEN

Beta-adrenergic blockade has been associated with improved cancer survival in patients with triple-negative breast cancer (TNBC), but the mechanisms of these effects remain unclear. In clinical epidemiological analyses, we identified a relationship between beta-blocker use and anthracycline chemotherapy in protecting against TNBC progression, disease recurrence, and mortality. We recapitulated the effect of beta-blockade on anthracycline efficacy in xenograft mouse models of TNBC. In metastatic 4T1.2 and MDA-MB-231 mouse models of TNBC, beta-blockade improved the efficacy of the anthracycline doxorubicin by reducing metastatic development. We found that anthracycline chemotherapy alone, in the absence of beta-blockade, increased sympathetic nerve fiber activity and norepinephrine concentration in mammary tumors through the induction of nerve growth factor (NGF) by tumor cells. Moreover, using preclinical models and clinical samples, we found that anthracycline chemotherapy up-regulated ß2-adrenoceptor expression and amplified receptor signaling in tumor cells. Neurotoxin inhibition of sympathetic neural signaling in mammary tumors using 6-hydroxydopamine or genetic deletion of NGF or ß2-adrenoceptor in tumor cells enhanced the therapeutic effect of anthracycline chemotherapy by reducing metastasis in xenograft mouse models. These findings reveal a neuromodulatory effect of anthracycline chemotherapy that undermines its potential therapeutic impact, which can be overcome by inhibiting ß2-adrenergic signaling in the tumor microenvironment. Supplementing anthracycline chemotherapy with adjunctive ß2-adrenergic antagonists represents a potential therapeutic strategy for enhancing the clinical management of TNBC.


Asunto(s)
Antraciclinas , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Antraciclinas/farmacología , Antraciclinas/uso terapéutico , Neoplasias de la Mama Triple Negativas/genética , Factor de Crecimiento Nervioso/uso terapéutico , Línea Celular Tumoral , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptores Adrenérgicos/uso terapéutico , Microambiente Tumoral
3.
Cell Rep ; 38(3): 110282, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35045295

RESUMEN

Although circular RNAs (circRNAs) are enriched in the brain, their relevance for brain function and psychiatric disorders is poorly understood. Here, we show that circHomer1 is inversely associated with relative HOMER1B mRNA isoform levels in both the orbitofrontal cortex (OFC) and stem-cell-derived neuronal cultures of subjects with psychiatric disorders. We further demonstrate that in vivo circHomer1 knockdown (KD) within the OFC can inhibit the synaptic expression of Homer1b mRNA. Furthermore, we show that circHomer1 directly binds to Homer1b mRNA and that Homer1b-specific KD increases synaptic circHomer1 levels and improves OFC-mediated behavioral flexibility. Importantly, double circHomer1 and Homer1b in vivo co-KD results in a complete rescue in circHomer1-associated alterations in both chance reversal learning and synaptic gene expression. Lastly, we uncover an RNA-binding protein that can directly bind to circHomer1 and promote its biogenesis. Taken together, our data provide mechanistic insights into the importance of circRNAs in brain function and disease.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas de Andamiaje Homer/metabolismo , Corteza Prefrontal/metabolismo , ARN Circular/metabolismo , Aprendizaje Inverso/fisiología , Animales , Trastorno Bipolar/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Neuron ; 106(6): 912-926.e5, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32304628

RESUMEN

Depression is a common disorder that affects women at twice the rate of men. Here, we report that long non-coding RNAs (lncRNAs), a recently discovered class of regulatory transcripts, represent about one-third of the differentially expressed genes in the brains of depressed humans and display complex region- and sex-specific patterns of regulation. We identified the primate-specific, neuronal-enriched gene LINC00473 as downregulated in prefrontal cortex (PFC) of depressed females but not males. Using viral-mediated gene transfer to express LINC00473 in adult mouse PFC neurons, we mirrored the human sex-specific phenotype by inducing stress resilience solely in female mice. This sex-specific phenotype was accompanied by changes in synaptic function and gene expression selectively in female mice and, along with studies of human neuron-like cells in culture, implicates LINC00473 as a CREB effector. Together, our studies identify LINC00473 as a female-specific driver of stress resilience that is aberrant in female depression.


Asunto(s)
Trastorno Depresivo Mayor/genética , Corteza Prefrontal/metabolismo , ARN Largo no Codificante/genética , Resiliencia Psicológica , Estrés Psicológico/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Conducta Animal , Depresión/genética , Depresión/metabolismo , Trastorno Depresivo Mayor/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Neuronas/metabolismo , ARN Largo no Codificante/metabolismo , RNA-Seq , Factores Sexuales , Estrés Psicológico/metabolismo , Adulto Joven
5.
Mol Psychiatry ; 25(11): 2712-2727, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31988434

RESUMEN

Although circular RNAs (circRNAs) are enriched in the mammalian brain, very little is known about their potential involvement in brain function and psychiatric disease. Here, we show that circHomer1a, a neuronal-enriched circRNA abundantly expressed in the frontal cortex, derived from Homer protein homolog 1 (HOMER1), is significantly reduced in both the prefrontal cortex (PFC) and induced pluripotent stem cell-derived neuronal cultures from patients with schizophrenia (SCZ) and bipolar disorder (BD). Moreover, alterations in circHomer1a were positively associated with the age of onset of SCZ in both the dorsolateral prefrontal cortex (DLPFC) and orbitofrontal cortex (OFC). No correlations between the age of onset of SCZ and linear HOMER1 mRNA were observed, whose expression was mostly unaltered in BD and SCZ postmortem brain. Using in vivo circRNA-specific knockdown of circHomer1a in mouse PFC, we show that it modulates the expression of numerous alternative mRNA transcripts from genes involved in synaptic plasticity and psychiatric disease. Intriguingly, in vivo circHomer1a knockdown in mouse OFC resulted in specific deficits in OFC-mediated cognitive flexibility. Lastly, we demonstrate that the neuronal RNA-binding protein HuD binds to circHomer1a and can influence its synaptic expression in the frontal cortex. Collectively, our data uncover a novel psychiatric disease-associated circRNA that regulates synaptic gene expression and cognitive flexibility.


Asunto(s)
Trastorno Bipolar/genética , Cognición , Regulación de la Expresión Génica , ARN Circular/genética , Esquizofrenia/genética , Sinapsis/metabolismo , Adulto , Animales , Femenino , Proteínas de Andamiaje Homer/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Corteza Prefrontal/metabolismo
6.
Nat Commun ; 9(1): 4926, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451900

RESUMEN

In the originally published version of this Article, the affiliation details for Eric E. Schadt and Radoslav Savic incorrectly omitted 'Sema4, a Mount Sinai venture, Stamford, Connecticut, USA'. This has been corrected in both the PDF and HTML versions of the Article.

7.
Nat Commun ; 9(1): 4412, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30356048

RESUMEN

A lack of biologically relevant screening models hinders the discovery of better treatments for schizophrenia (SZ) and other neuropsychiatric disorders. Here we compare the transcriptional responses of 8 commonly used cancer cell lines (CCLs) directly with that of human induced pluripotent stem cell (hiPSC)-derived neural progenitor cells (NPCs) from 12 individuals with SZ and 12 controls across 135 drugs, generating 4320 unique drug-response transcriptional signatures. We identify those drugs that reverse post-mortem SZ-associated transcriptomic signatures, several of which also differentially regulate neuropsychiatric disease-associated genes in a cell type (hiPSC NPC vs. CCL) and/or a diagnosis (SZ vs. control)-dependent manner. Overall, we describe a proof-of-concept application of transcriptomic drug screening to hiPSC-based models, demonstrating that the drug-induced gene expression differences observed with patient-derived hiPSC NPCs are enriched for SZ biology, thereby revealing a major advantage of incorporating cell type and patient-specific platforms in drug discovery.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Esquizofrenia/metabolismo , Línea Celular , Línea Celular Tumoral , Dimetilsulfóxido/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Control de Calidad , Transcriptoma
8.
Nat Commun ; 8(1): 2225, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29263384

RESUMEN

The power of human induced pluripotent stem cell (hiPSC)-based studies to resolve the smaller effects of common variants within the size of cohorts that can be realistically assembled remains uncertain. We identified and accounted for a variety of technical and biological sources of variation in a large case/control schizophrenia (SZ) hiPSC-derived cohort of neural progenitor cells and neurons. Reducing the stochastic effects of the differentiation process by correcting for cell type composition boosted the SZ signal and increased the concordance with post-mortem data sets. We predict a growing convergence between hiPSC and post-mortem studies as both approaches expand to larger cohort sizes. For studies of complex genetic disorders, to maximize the power of hiPSC cohorts currently feasible, in most cases and whenever possible, we recommend expanding the number of individuals even at the expense of the number of replicate hiPSC clones.


Asunto(s)
Encéfalo/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , ARN Mensajero/metabolismo , Esquizofrenia/genética , Adolescente , Adulto , Antígenos de Superficie/genética , Autopsia , Estudios de Casos y Controles , Niño , Variaciones en el Número de Copia de ADN , Femenino , Humanos , Modelos Lineales , Masculino , Proteína Homeótica Nanog/genética , Nestina/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteoglicanos/genética , Factores de Transcripción SOXB1/genética , Análisis de Secuencia de ARN , Antígenos Embrionarios Específico de Estadio/genética , Sinapsinas/genética , Transcriptoma , Adulto Joven
10.
Stem Cell Reports ; 9(2): 615-628, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28757163

RESUMEN

Modulation of transcription, either synthetic activation or repression, via dCas9-fusion proteins is a relatively new methodology with the potential to facilitate high-throughput up- or downregulation studies of gene function. Genetic studies of neurodevelopmental disorders have identified a growing list of risk variants, including both common single-nucleotide variants and rare copy-number variations, many of which are associated with genes having limited functional annotations. By applying a CRISPR-mediated gene-activation/repression platform to populations of human-induced pluripotent stem cell-derived neural progenitor cells, neurons, and astrocytes, we demonstrate that it is possible to manipulate endogenous expression levels of candidate neuropsychiatric risk genes across these three cell types. Although proof-of-concept studies using catalytically inactive Cas9-fusion proteins to modulate transcription have been reported, here we present a detailed survey of the reproducibility of gRNA positional effects across a variety of neurodevelopmental disorder-relevant risk genes, donors, neural cell types, and dCas9 effectors.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Imagen Molecular , Transcriptoma
11.
Stem Cell Reports ; 9(2): 600-614, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28757165

RESUMEN

Growing evidence implicates the importance of glia, particularly astrocytes, in neurological and psychiatric diseases. Here, we describe a rapid and robust method for the differentiation of highly pure populations of replicative astrocytes from human induced pluripotent stem cells (hiPSCs), via a neural progenitor cell (NPC) intermediate. We evaluated this protocol across 42 NPC lines (derived from 30 individuals). Transcriptomic analysis demonstrated that hiPSC-astrocytes from four individuals are highly similar to primary human fetal astrocytes and characteristic of a non-reactive state. hiPSC-astrocytes respond to inflammatory stimulants, display phagocytic capacity, and enhance microglial phagocytosis. hiPSC-astrocytes also possess spontaneous calcium transient activity. Our protocol is a reproducible, straightforward (single medium), and rapid (<30 days) method to generate populations of hiPSC-astrocytes that can be used for neuron-astrocyte and microglia-astrocyte co-cultures for the study of neuropsychiatric disorders.


Asunto(s)
Astrocitos/citología , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Astrocitos/metabolismo , Calcio/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Células Cultivadas , Biología Computacional/métodos , Citocinas/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Microglía/inmunología , Microglía/metabolismo , Anotación de Secuencia Molecular , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fagocitosis , Transcriptoma
12.
Cell Stem Cell ; 21(2): 274-283.e5, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28736217

RESUMEN

Zika virus (ZIKV) infects fetal and adult human brain and is associated with serious neurological complications. To date, no therapeutic treatment is available to treat ZIKV-infected patients. We performed a high-content chemical screen using human pluripotent stem cell-derived cortical neural progenitor cells (hNPCs) and found that hippeastrine hydrobromide (HH) and amodiaquine dihydrochloride dihydrate (AQ) can inhibit ZIKV infection in hNPCs. Further validation showed that HH also rescues ZIKV-induced growth and differentiation defects in hNPCs and human fetal-like forebrain organoids. Finally, HH and AQ inhibit ZIKV infection in adult mouse brain in vivo. Strikingly, HH suppresses viral propagation when administered to adult mice with active ZIKV infection, highlighting its therapeutic potential. Our approach highlights the power of stem cell-based screens and validation in human forebrain organoids and mouse models in identifying drug candidates for treating ZIKV infection and related neurological complications in fetal and adult patients.


Asunto(s)
Antivirales/uso terapéutico , Encéfalo/virología , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Organoides/virología , Infección por el Virus Zika/tratamiento farmacológico , Virus Zika/fisiología , Adolescente , Alcaloides de Amaryllidaceae/farmacología , Amodiaquina/farmacología , Animales , Antivirales/farmacología , Línea Celular , Niño , Femenino , Feto/efectos de los fármacos , Feto/virología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones SCID , Células-Madre Neurales/efectos de los fármacos , Organoides/efectos de los fármacos , Virus Zika/efectos de los fármacos , Infección por el Virus Zika/patología
13.
Nat Genet ; 49(8): 1239-1250, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28671686

RESUMEN

We report locus-specific disintegration of megabase-scale chromosomal conformations in brain after neuronal ablation of Setdb1 (also known as Kmt1e; encodes a histone H3 lysine 9 methyltransferase), including a large topologically associated 1.2-Mb domain conserved in humans and mice that encompasses >70 genes at the clustered protocadherin locus (hereafter referred to as cPcdh). The cPcdh topologically associated domain (TADcPcdh) in neurons from mutant mice showed abnormal accumulation of the transcriptional regulator and three-dimensional (3D) genome organizer CTCF at cryptic binding sites, in conjunction with DNA cytosine hypomethylation, histone hyperacetylation and upregulated expression. Genes encoding stochastically expressed protocadherins were transcribed by increased numbers of cortical neurons, indicating relaxation of single-cell constraint. SETDB1-dependent loop formations bypassed 0.2-1 Mb of linear genome and radiated from the TADcPcdh fringes toward cis-regulatory sequences within the cPcdh locus, counterbalanced shorter-range facilitative promoter-enhancer contacts and carried loop-bound polymorphisms that were associated with genetic risk for schizophrenia. We show that the SETDB1 repressor complex, which involves multiple KRAB zinc finger proteins, shields neuronal genomes from excess CTCF binding and is critically required for structural maintenance of TADcPcdh.


Asunto(s)
Cromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Neuronas/metabolismo , Animales , Factor de Unión a CCCTC , Cadherinas/genética , Línea Celular , Metilación de ADN , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Masculino , Ratones , Mutación , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , Proteínas Represoras/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-28149327

RESUMEN

BACKGROUND: Intrauterine exposure to maternal smoking is linked to impaired executive function and behavioral problems in the offspring. Maternal smoking is associated with reduced fetal brain growth and smaller volume of cortical gray matter in childhood, indicating that prenatal exposure to tobacco may impact cortical development and manifest as behavioral problems. Cellular development is mediated by changes in epigenetic modifications such as DNA methylation, which can be affected by exposure to tobacco. RESULTS: In this study, we sought to ascertain how maternal smoking during pregnancy affects global DNA methylation profiles of the developing dorsolateral prefrontal cortex (DLPFC) during the second trimester of gestation. When DLPFC methylation profiles (assayed via Illumina, HM450) of smoking-exposed and unexposed fetuses were compared, no differentially methylated regions (DMRs) passed the false discovery correction (FDR ≤ 0.05). However, the most significant DMRs were hypomethylated CpG Islands within the promoter regions of GNA15 and SDHAP3 of smoking-exposed fetuses. Interestingly, the developmental up-regulation of SDHAP3 mRNA was delayed in smoking-exposed fetuses. Interaction analysis between gestational age and smoking exposure identified significant DMRs annotated to SYCE3, C21orf56/LSS, SPAG1 and RNU12/POLDIP3 that passed FDR. Furthermore, utilizing established methods to estimate cell proportions by DNA methylation, we found that exposed DLPFC samples contained a lower proportion of neurons in samples from fetuses exposed to maternal smoking. We also show through in vitro experiments that nicotine impedes the differentiation of neurons independent of cell death. CONCLUSIONS: We found evidence that intrauterine smoking exposure alters the developmental patterning of DNA methylation and gene expression and is associated with reduced mature neuronal content, effects that are likely driven by nicotine.


Asunto(s)
Encéfalo/metabolismo , Metilación de ADN , Exposición Materna , Fumar , Encéfalo/patología , Femenino , Desarrollo Fetal/genética , Feto/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Edad Gestacional , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Embarazo , Segundo Trimestre del Embarazo , Regiones Promotoras Genéticas , Succinato Deshidrogenasa/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
15.
Neurochem Int ; 106: 85-93, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27744003

RESUMEN

Human induced pluripotent stem cells (hiPSCs) can theoretically yield limitless supplies of cells fated to any cell type that comprise the human organism, making them a new tool by which to potentially overcome caveats in current biomedical research. In vitro derivation of central nervous system (CNS) cell types has the potential to provide material for drug discovery and validation, safety and toxicity assays, cell replacement therapy and the elucidation of previously unknown disease mechanisms. However, current two-dimensional (2D) CNS differentiation protocols do not faithfully recapitulate the spatial organization of heterogeneous tissue, nor the cell-cell interactions, cell-extracellular matrix interactions, or specific physiological functions generated within complex tissue such as the brain. In an effort to overcome 2D protocol limitations, there have been advancements in deriving highly complicated 3D neural organoid structures. Herein we provide a synopsis of the derivation and application of neural organoids and discuss recent advancements and remaining challenges on the full potential of this novel technological platform.


Asunto(s)
Biología Evolutiva/tendencias , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Organoides/efectos de los fármacos , Organoides/fisiología , Fenotipo , Animales , Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/tendencias , Biología Evolutiva/métodos , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/tendencias , Humanos , Preparaciones Farmacéuticas/administración & dosificación , Pruebas de Toxicidad/métodos , Pruebas de Toxicidad/tendencias
16.
Cell Rep ; 15(5): 1024-1036, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-27117414

RESUMEN

Converging evidence indicates that microRNAs (miRNAs) may contribute to disease risk for schizophrenia (SZ). We show that microRNA-9 (miR-9) is abundantly expressed in control neural progenitor cells (NPCs) but also significantly downregulated in a subset of SZ NPCs. We observed a strong correlation between miR-9 expression and miR-9 regulatory activity in NPCs as well as between miR-9 levels/activity, neural migration, and diagnosis. Overexpression of miR-9 was sufficient to ameliorate a previously reported neural migration deficit in SZ NPCs, whereas knockdown partially phenocopied aberrant migration in control NPCs. Unexpectedly, proteomic- and RNA sequencing (RNA-seq)-based analysis revealed that these effects were mediated primarily by small changes in expression of indirect miR-9 targets rather than large changes in direct miR-9 targets; these indirect targets are enriched for migration-associated genes. Together, these data indicate that aberrant levels and activity of miR-9 may be one of the many factors that contribute to SZ risk, at least in a subset of patients.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Células-Madre Neurales/metabolismo , Esquizofrenia/genética , Esquizofrenia/patología , Estudios de Casos y Controles , Movimiento Celular/genética , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/metabolismo , Modelos Biológicos , Anotación de Secuencia Molecular , Células-Madre Neurales/patología , Neuronas/metabolismo , Proteoma/metabolismo , Factores de Transcripción/metabolismo
17.
Methods ; 101: 113-24, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26626326

RESUMEN

Since the discovery of somatic reprogramming, human induced pluripotent stem cells (hiPSCs) have been exploited to model a variety of neurological and psychiatric disorders. Because hiPSCs represent an almost limitless source of patient-derived neurons that retain the genetic variations thought to contribute to disease etiology, they have been heralded as a patient-specific platform for high throughput drug screening. However, the utility of current protocols for generating neurons from hiPSCs remains limited by protracted differentiation timelines and heterogeneity of the neuronal phenotypes produced. Neuronal induction via the forced expression of exogenous transcription factors rapidly induces defined populations of functional neurons from fibroblasts and hiPSCs. Here, we describe an adapted protocol that accelerates maturation of functional excitatory neurons from hiPSC-derived neural progenitor cells (NPCs) via lentiviral transduction of Neurogenin 2 (using both mNgn2 and hNGN2). This methodology, relying upon a robust and scalable starting population of hiPSC NPCs, should be readily amenable to scaling for hiPSC-based high-throughput drug screening.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Proteínas del Tejido Nervioso/fisiología , Técnicas de Cultivo de Célula , Células Cultivadas , Reprogramación Celular , Expresión Génica , Humanos , Lentivirus , Células-Madre Neurales/fisiología , Activación Transcripcional
18.
Front Cell Neurosci ; 9: 104, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25873861

RESUMEN

PITX3 expression is confined to adult midbrain dopaminergic (mDA) neurons. In this study we describe the generation and basic functional characteristics of mDA neurons derived from a human pluripotent stem cell (hPSC) line expressing eGFP under the control of the PITX3 promoter. Flow cytometry showed that eGFP was evident in 15% of the neuron population at day 12 of differentiation and this level was maintained until at least day 80. From days 20 to 80 of differentiation intracellular chloride decreased and throughout this period around ∼20% of PITX3(eGFP/w) neurons exhibited spontaneous Ca(2+) transients (from 3.3 ± 0.3 to 5.0 ± 0.1 min(-1), respectively). These neurons also responded to any of ATP, glutamate, acetylcholine, or noradrenaline with elevations of intracellular calcium. As neuronal cultures matured more dopamine was released and single PITX3(eGFP/w) neurons began to respond to more than one neurotransmitter. MPP(+) and tumor necrosis factor (TNF), but not prostaglandin E2, caused death of the ∼50% of PITX3(eGFP/w) neurons (day 80). Tracking eGFP using time lapse confocal microscopy over 24 h demonstrated significant TNF-mediated neurite retraction over time. This work now shows that these PITX3(eGFP/w) neurons are amenable to flow cytometry, release dopamine and respond to multiple neurotransmitters with elevations of intracellular calcium, we believe that they represent a versatile system for neuropharmacological and neurotoxicological studies.

19.
NPJ Schizophr ; 12015 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-26985448

RESUMEN

Neurodevelopmental disorders, such as autism spectrum disorders (ASD) and schizophrenia (SZ), are complex disorders with a high degree of heritability. Genetic studies have identified several candidate genes associated with these disorders, including contactin-associated protein-like 2 (CNTNAP2). Traditionally, in animal models or in vitro, the function of CNTNAP2 has been studied by genetic deletion or transcriptional knockdown, which reduce the expression of the entire gene; however, it remains unclear whether the mutations identified in clinical settings are sufficient to alter CNTNAP2 expression in human neurons. Here, using human induced pluripotent stem cells (hiPSCs) derived from two individuals with a large (289kb) and heterozygous deletion in CNTNAP2 (affecting exons 14-15) and discordant clinical outcomes, we have characterized CNTNAP2 expression patterns in hiPSC neural progenitor cells (NPCs), two independent populations of hiPSC-derived neurons and hiPSC-derived oligodendrocyte precursor cells (OPCs). First, we observed exon-specific changes in CNTNAP2 expression in both carriers; although the expression of exons 14-15 is significantly decreased, the expression of other exons is upregulated. Second, we observed significant differences in patterns of allele-specific expression in CNTNAP2 carriers that were consistent with clinical outcome. Third, we observed a robust neural migration phenotype that correlated with diagnosis and exon- and allele-specific CNTNAP2 expression patterns, but not with genotype. In all, our data highlight the importance of considering the nature, location and regulation of mutated alleles when attempting to connect GWAS studies to gene function.

20.
J Vis Exp ; (90): e51764, 2014 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-25177806

RESUMEN

One major limitation with current human embryonic stem cell (ESC) differentiation protocols is the generation of heterogeneous cell populations. These cultures contain the cells of interest, but are also contaminated with undifferentiated ESCs, non-neural derivatives and other neuronal subtypes. This limits their use in in vitro and in vivo applications, such as in vitro modeling for drug discovery or cell replacement therapy. To help overcome this, reporter cell lines, which offer a means to visualize, track and isolate cells of interest, can be engineered. However, to achieve this in human embryonic stem cells via conventional homologous recombination is extremely inefficient. This protocol describes targeting of the Pituitary homeobox 3 (PITX3) locus in human embryonic stem cells using custom designed zinc-finger nucleases, which introduce site-specific double-strand DNA breaks, together with a PITX3-EGFP-specific DNA donor vector. Following the generation of the PITX3 reporter cell line, it can then be differentiated using published protocols for use in studies such as in vitro Parkinson's disease modeling or cell replacement therapy.


Asunto(s)
Desoxirribonucleasas/metabolismo , Células Madre Embrionarias/fisiología , Marcación de Gen/métodos , Dedos de Zinc , Animales , Rastreo Celular , Roturas del ADN de Doble Cadena , Desoxirribonucleasas/química , Electroporación/métodos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Proteínas Recombinantes de Fusión/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA