Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Commun ; 6(3): fcae202, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38911266

RESUMEN

While voltage-gated potassium channels have critical roles in controlling neuronal excitability, they also have non-ion-conducting functions. Kv8.1, encoded by the KCNV1 gene, is a 'silent' ion channel subunit whose biological role is complex since Kv8.1 subunits do not form functional homotetramers but assemble with Kv2 to modify its ion channel properties. We profiled changes in ion channel expression in amyotrophic lateral sclerosis patient-derived motor neurons carrying a superoxide dismutase 1(A4V) mutation to identify what drives their hyperexcitability. A major change identified was a substantial reduction of KCNV1/Kv8.1 expression, which was also observed in patient-derived neurons with C9orf72 expansion. We then studied the effect of reducing KCNV1/Kv8.1 expression in healthy motor neurons and found it did not change neuronal firing but increased vulnerability to cell death. A transcriptomic analysis revealed dysregulated metabolism and lipid/protein transport pathways in KCNV1/Kv8.1-deficient motor neurons. The increased neuronal vulnerability produced by the loss of KCNV1/Kv8.1 was rescued by knocking down Kv2.2, suggesting a potential Kv2.2-dependent downstream mechanism in cell death. Our study reveals, therefore, unsuspected and distinct roles of Kv8.1 and Kv2.2 in amyotrophic lateral sclerosis-related neurodegeneration.

2.
iScience ; 26(5): 106701, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37207277

RESUMEN

Genetics have nominated many schizophrenia risk genes and identified convergent signals between schizophrenia and neurodevelopmental disorders. However, functional interpretation of the nominated genes in the relevant brain cell types is often lacking. We executed interaction proteomics for six schizophrenia risk genes that have also been implicated in neurodevelopment in human induced cortical neurons. The resulting protein network is enriched for common variant risk of schizophrenia in Europeans and East Asians, is down-regulated in layer 5/6 cortical neurons of individuals affected by schizophrenia, and can complement fine-mapping and eQTL data to prioritize additional genes in GWAS loci. A sub-network centered on HCN1 is enriched for common variant risk and contains proteins (HCN4 and AKAP11) enriched for rare protein-truncating mutations in individuals with schizophrenia and bipolar disorder. Our findings showcase brain cell-type-specific interactomes as an organizing framework to facilitate interpretation of genetic and transcriptomic data in schizophrenia and its related disorders.

3.
Cell Genom ; 3(3): 100250, 2023 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-36950384

RESUMEN

Autism spectrum disorders (ASDs) have been linked to genes with enriched expression in the brain, but it is unclear how these genes converge into cell-type-specific networks. We built a protein-protein interaction network for 13 ASD-associated genes in human excitatory neurons derived from induced pluripotent stem cells (iPSCs). The network contains newly reported interactions and is enriched for genetic and transcriptional perturbations observed in individuals with ASDs. We leveraged the network data to show that the ASD-linked brain-specific isoform of ANK2 is important for its interactions with synaptic proteins and to characterize a PTEN-AKAP8L interaction that influences neuronal growth. The IGF2BP1-3 complex emerged as a convergent point in the network that may regulate a transcriptional circuit of ASD-associated genes. Our findings showcase cell-type-specific interactomes as a framework to complement genetic and transcriptomic data and illustrate how both individual and convergent interactions can lead to biological insights into ASDs.

4.
Mol Ther ; 30(8): 2646-2663, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35690906

RESUMEN

On August 18, 2021, the American Society of Gene and Cell Therapy (ASGCT) hosted a virtual roundtable on adeno-associated virus (AAV) integration, featuring leading experts in preclinical and clinical AAV gene therapy, to further contextualize and understand this phenomenon. Recombinant AAV (rAAV) vectors are used to develop therapies for many conditions given their ability to transduce multiple cell types, resulting in long-term expression of transgenes. Although most rAAV DNA typically remains episomal, some rAAV DNA becomes integrated into genomic DNA at a low frequency, and rAAV insertional mutagenesis has been shown to lead to tumorigenesis in neonatal mice. Currently, the risk of rAAV-mediated oncogenesis in humans is theoretical because no confirmed genotoxic events have been reported to date. However, because insertional mutagenesis has been reported in a small number of murine studies, there is a need to characterize this genotoxicity to inform research, regulatory needs, and patient care. The purpose of this white paper is to review the evidence of rAAV-related host genome integration in animal models and possible risks of insertional mutagenesis in patients. In addition, technical considerations, regulatory guidance, and bioethics are discussed.


Asunto(s)
Dependovirus , Vectores Genéticos , Animales , Dependovirus/genética , Vectores Genéticos/genética , Humanos , Ratones , Mutagénesis Insercional , Plásmidos , Transgenes , Integración Viral
5.
Nat Commun ; 12(1): 2580, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972534

RESUMEN

Combining genetic and cell-type-specific proteomic datasets can generate biological insights and therapeutic hypotheses, but a technical and statistical framework for such analyses is lacking. Here, we present an open-source computational tool called Genoppi (lagelab.org/genoppi) that enables robust, standardized, and intuitive integration of quantitative proteomic results with genetic data. We use Genoppi to analyze 16 cell-type-specific protein interaction datasets of four proteins (BCL2, TDP-43, MDM2, PTEN) involved in cancer and neurological disease. Through systematic quality control of the data and integration with published protein interactions, we show a general pattern of both cell-type-independent and cell-type-specific interactions across three cancer cell types and one human iPSC-derived neuronal cell type. Furthermore, through the integration of proteomic and genetic datasets in Genoppi, our results suggest that the neuron-specific interactions of these proteins are mediating their genetic involvement in neurodegenerative diseases. Importantly, our analyses suggest that human iPSC-derived neurons are a relevant model system for studying the involvement of BCL2 and TDP-43 in amyotrophic lateral sclerosis.


Asunto(s)
Biología Computacional/métodos , Estudio de Asociación del Genoma Completo/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neuronas/metabolismo , Programas Informáticos , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genómica , Humanos , Mutación , Polimorfismo de Nucleótido Simple , Unión Proteica , Proteómica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Espectrometría de Masas en Tándem
6.
Mol Autism ; 12(1): 10, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33557935

RESUMEN

Autism spectrum disorder (ASD) comprises a group of neurodevelopmental disorders characterized by impaired social interactions as well as the presentation of restrictive and repetitive behaviors. ASD is highly heritable but genetically heterogenous with both common and rare genetic variants collaborating to predispose individuals to the disorder. In this review, we synthesize recent efforts to develop human induced pluripotent stem cell (iPSC)-derived models of ASD-related phenotypes. We firstly address concerns regarding the relevance and validity of available neuronal iPSC-derived models. We then critically evaluate the robustness of various differentiation and cell culture protocols used for producing cell types of relevance to ASD. By exploring iPSC models of ASD reported thus far, we examine to what extent cellular and neuronal phenotypes with potential relevance to ASD can be linked to genetic variants found to underlie it. Lastly, we outline promising strategies by which iPSC technology can both enhance the power of genetic studies to identify ASD risk factors and nominate pathways that are disrupted across groups of ASD patients that might serve as common points for therapeutic intervention.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Células Madre Pluripotentes Inducidas/metabolismo , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/terapia , Biomarcadores , Técnicas de Cultivo de Célula , Estudios de Asociación Genética/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Terapia Molecular Dirigida , Herencia Multifactorial , Fenotipo
7.
Bio Protoc ; 10(17): e3748, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-33659408

RESUMEN

The efficiency of cleavage of individual CRISPR/Cas9-sgRNAs remains difficult to predict based on the CRISPR target sequence alone. Different intracellular environments (dependent on cell type or cell cycle state for example) may affect sgRNA efficiency by altering accessibility of genomic DNA through DNA modifications such as epigenetic marks and DNA-binding proteins (e.g., histones) as well as alteration of the chromatin state of genomic DNA within the nucleus. We recently reported a multi-step screening method for the identification of efficient sgRNAs targeting the Herpes simplex virus (HSV-1) genome and reported a differential mechanism for viral inhibition by CRISPR-Cas9 in the latent versus lytic phase. The screening platform detailed in this protocol allows step-by-step testing of the efficiency of cleavage in a cell-free system and in the context of viral target cells such as human foreskin fibroblasts followed by functional testing of the effects of CRISPR/sgRNA on viral protein expression, replication, and reactivation. This strategy could be readily applied to other target cells such as pluripotent stem cell-derived human sensory neurons or other human DNA viruses.

8.
Elife ; 82019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31789594

RESUMEN

Herpes simplex virus (HSV) establishes lifelong latent infection and can cause serious human disease, but current antiviral therapies target lytic but not latent infection. We screened for sgRNAs that cleave HSV-1 DNA sequences efficiently in vitro and used these sgRNAs to observe the first editing of quiescent HSV-1 DNA. The sgRNAs targeted lytic replicating viral DNA genomes more efficiently than quiescent genomes, consistent with the open structure of lytic chromatin. Editing of latent genomes caused short indels while editing of replicating genomes produced indels, linear molecules, and large genomic sequence loss around the gRNA target site. The HSV ICP0 protein and viral DNA replication increased the loss of DNA sequences around the gRNA target site. We conclude that HSV, by promoting open chromatin needed for viral gene expression and by inhibiting the DNA damage response, makes the genome vulnerable to a novel form of editing by CRISPR-Cas9 during lytic replication.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Edición Génica , Genes Virales , Herpesviridae/genética , Animales , Secuencia de Bases , Línea Celular , Reparación del ADN/genética , ADN Viral/genética , Regulación Viral de la Expresión Génica , Herpesvirus Humano 1/genética , Humanos , Modelos Genéticos , Mutagénesis/genética , ARN Guía de Kinetoplastida/genética , Replicación Viral/genética
9.
Nat Neurosci ; 22(2): 229-242, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30664768

RESUMEN

We generated cortical interneurons (cINs) from induced pluripotent stem cells derived from 14 healthy controls and 14 subjects with schizophrenia. Both healthy control cINs and schizophrenia cINs were authentic, fired spontaneously, received functional excitatory inputs from host neurons, and induced GABA-mediated inhibition in host neurons in vivo. However, schizophrenia cINs had dysregulated expression of protocadherin genes, which lie within documented schizophrenia loci. Mice lacking protocadherin-α showed defective arborization and synaptic density of prefrontal cortex cINs and behavioral abnormalities. Schizophrenia cINs similarly showed defects in synaptic density and arborization that were reversed by inhibitors of protein kinase C, a downstream kinase in the protocadherin pathway. These findings reveal an intrinsic abnormality in schizophrenia cINs in the absence of any circuit-driven pathology. They also demonstrate the utility of homogenous and functional populations of a relevant neuronal subtype for probing pathogenesis mechanisms during development.


Asunto(s)
Cadherinas/metabolismo , Interneuronas/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/metabolismo , Transducción de Señal/fisiología , Animales , Cadherinas/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas , Interneuronas/patología , Masculino , Ratones , Ratones Noqueados , Corteza Prefrontal/patología , Protocadherinas , Esquizofrenia/patología , Sinapsis/genética , Sinapsis/metabolismo
10.
J Assist Reprod Genet ; 35(10): 1763-1771, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30120633

RESUMEN

PURPOSE: To provide a commentary on our understanding of the role that the Hippo signaling pathway may play in patients with polycystic ovarian syndrome (PCOS) and how this understanding may impact the diagnosis of PCOS. METHODS: We assessed publications discussing the role of the Hippo signaling pathway in the ovary. In particular, we discuss how Hippo signaling disruption after ovarian fragmentation, combined with treating ovarian fragments with phosphatase and tensin homolog (PTEN) inhibitors and phosphoinositide-3-kinase stimulators to augment AKT signaling, has been used in treatment of patients with primary ovarian insufficiency. Furthermore, we discuss our own data on variations in Hippo signaling pathway gene expression in cumulus cells isolated from women undergoing IVF with a previous diagnosis of PCOS. RESULTS AND CONCLUSIONS: Aberrant Hippo signaling in PCOS patients is likely a contributing mechanism to the multifactorial etiology of the disease. Given the challenge of discerning the underlying etiology of oligo-ovulation in some patients, especially those with normal body mass indices, and the need for customized stimulation protocols for PCOS patients who have an increased risk of over-response and higher percentage of immature oocyte yield, it is important to identify these patients prior to treatment. Hippo gene expression fingerprints could potentially be used to more accurately define patients with PCOS. Additionally, targeting this pathway with pharmacologic agents could lead to non-surgical therapeutic options for PCOS.


Asunto(s)
Fertilización In Vitro , Ovario/metabolismo , Síndrome del Ovario Poliquístico/genética , Proteínas Serina-Treonina Quinasas/genética , Femenino , Vía de Señalización Hippo , Humanos , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Transducción de Señal
11.
Mol Biol Cell ; 25(17): 2571-8, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25009283

RESUMEN

Mutations in the RNA-binding protein FUS have been shown to cause the neurodegenerative disease amyotrophic lateral sclerosis (ALS). We investigate whether mutant FUS protein in ALS patient-derived fibroblasts affects normal FUS functions in the nucleus. We investigated fibroblasts from two ALS patients possessing different FUS mutations and a normal control. Fibroblasts from these patients have their nuclear FUS protein trapped in SDS-resistant aggregates. Genome-wide analysis reveals an inappropriate accumulation of Ser-2 phosphorylation on RNA polymerase II (RNA Pol II) near the transcription start sites of 625 genes for ALS patient cells and after small interfering RNA (siRNA) knockdown of FUS in normal fibroblasts. Furthermore, both the presence of mutant FUS protein and siRNA knockdown of wild-type FUS correlate with altered distribution of RNA Pol II within fibroblast nuclei. A loss of FUS function in orchestrating Ser-2 phosphorylation of the CTD of RNA Pol II is detectable in ALS patient-derived fibroblasts expressing mutant FUS protein, even when the FUS protein remains largely nuclear. A likely explanation for this loss of function is the aggregation of FUS protein in nuclei. Thus our results suggest a specific mechanism by which mutant FUS can have biological consequences other than by the formation of cytoplasmic aggregates.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Núcleo Celular/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Esclerosis Amiotrófica Lateral/genética , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Fosforilación , Agregado de Proteínas , Interferencia de ARN , ARN Polimerasa II/metabolismo , Proteína FUS de Unión a ARN/antagonistas & inhibidores , Proteína FUS de Unión a ARN/genética , Sitio de Iniciación de la Transcripción
12.
Neuron ; 81(3): 536-543, 2014 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-24507191

RESUMEN

The RNA-binding protein TDP-43 regulates RNA metabolism at multiple levels, including transcription, RNA splicing, and mRNA stability. TDP-43 is a major component of the cytoplasmic inclusions characteristic of amyotrophic lateral sclerosis and some types of frontotemporal lobar degeneration. The importance of TDP-43 in disease is underscored by the fact that dominant missense mutations are sufficient to cause disease, although the role of TDP-43 in pathogenesis is unknown. Here we show that TDP-43 forms cytoplasmic mRNP granules that undergo bidirectional, microtubule-dependent transport in neurons in vitro and in vivo and facilitate delivery of target mRNA to distal neuronal compartments. TDP-43 mutations impair this mRNA transport function in vivo and in vitro, including in stem cell-derived motor neurons from ALS patients bearing any one of three different TDP-43 ALS-causing mutations. Thus, TDP-43 mutations that cause ALS lead to partial loss of a novel cytoplasmic function of TDP-43.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Transporte Axonal/genética , Proteínas de Unión al ADN/genética , Neuronas Motoras/metabolismo , Mutación/genética , ARN Mensajero/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Animales Modificados Genéticamente , Células Cultivadas , Corteza Cerebral/citología , Drosophila , Proteínas de Drosophila/genética , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Luminiscentes/genética , Ratones , Mitocondrias/metabolismo , Neuronas Motoras/ultraestructura , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
13.
Nature ; 495(7442): 474-80, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23474986

RESUMEN

CLP1 was the first mammalian RNA kinase to be identified. However, determining its in vivo function has been elusive. Here we generated kinase-dead Clp1 (Clp1(K/K)) mice that show a progressive loss of spinal motor neurons associated with axonal degeneration in the peripheral nerves and denervation of neuromuscular junctions, resulting in impaired motor function, muscle weakness, paralysis and fatal respiratory failure. Transgenic rescue experiments show that CLP1 functions in motor neurons. Mechanistically, loss of CLP1 activity results in accumulation of a novel set of small RNA fragments, derived from aberrant processing of tyrosine pre-transfer RNA. These tRNA fragments sensitize cells to oxidative-stress-induced p53 (also known as TRP53) activation and p53-dependent cell death. Genetic inactivation of p53 rescues Clp1(K/K) mice from the motor neuron loss, muscle denervation and respiratory failure. Our experiments uncover a mechanistic link between tRNA processing, formation of a new RNA species and progressive loss of lower motor neurons regulated by p53.


Asunto(s)
Neuronas Motoras/metabolismo , Neuronas Motoras/patología , ARN de Transferencia de Tirosina/metabolismo , Factores de Transcripción/metabolismo , Esclerosis Amiotrófica Lateral , Animales , Animales Recién Nacidos , Axones/metabolismo , Axones/patología , Muerte Celular , Diafragma/inervación , Pérdida del Embrión , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Exones/genética , Femenino , Fibroblastos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Atrofia Muscular Espinal , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/patología , Estrés Oxidativo , Procesamiento Postranscripcional del ARN , ARN de Transferencia de Tirosina/genética , Proteínas de Unión al ARN , Respiración , Nervios Espinales/citología , Factores de Transcripción/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Tirosina/genética , Tirosina/metabolismo
15.
Neuron ; 70(4): 626-44, 2011 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-21609821

RESUMEN

Among the disciplines of medicine, the study of neurological disorders is particularly challenging. The fundamental inaccessibility of the human neural types affected by disease prevents their isolation for in vitro studies of degenerative mechanisms or for drug screening efforts. However, the ability to reprogram readily accessible tissue from patients into pluripotent stem (iPS) cells may now provide a general solution to this shortage of human neurons. Gradually improving methods for directing the differentiation of patient-specific stem cells has enabled the production of several neural cell types affected by disease. Furthermore, initial studies with stem cell lines derived from individuals with pediatric, monogenic disorders have validated the stem cell approach to disease modeling, allowing relevant neural phenotypes to be observed and studied. Whether iPS cell-derived neurons will always faithfully recapitulate the same degenerative processes observed in patients and serve as platforms for drug discovery relevant to common late-onset diseases remains to be determined.


Asunto(s)
Modelos Teóricos , Enfermedades del Sistema Nervioso/cirugía , Trasplante de Células Madre/métodos , Células Madre , Animales , Diferenciación Celular/fisiología , Humanos , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/patología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Células Madre/citología , Células Madre/fisiología
16.
Cell Stem Cell ; 3(6): 637-48, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19041780

RESUMEN

It has been proposed that human embryonic stem cells could be used to provide an inexhaustible supply of differentiated cell types for the study of disease processes. Although methods for differentiating embryonic stem cells into specific cell types have become increasingly sophisticated, the utility of the resulting cells for modeling disease has not been determined. We have asked whether specific neuronal subtypes produced from human embryonic stem cells can be used to investigate the mechanisms leading to neural degeneration in amyotrophic lateral sclerosis (ALS). We show that human spinal motor neurons, but not interneurons, are selectively sensitive to the toxic effect of glial cells carrying an ALS-causing mutation in the SOD1 gene. Our findings demonstrate the relevance of these non-cell-autonomous effects to human motor neurons and more broadly demonstrate the utility of human embryonic stem cells for studying disease and identifying potential therapeutics.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Células Madre Embrionarias/metabolismo , Biología Molecular/métodos , Neuronas Motoras/metabolismo , Degeneración Nerviosa/metabolismo , Neuroglía/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Animales Recién Nacidos , Comunicación Celular/genética , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Línea Celular , Supervivencia Celular/genética , Células Cultivadas , Técnicas de Cocultivo , Células Madre Embrionarias/citología , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Neuronas Motoras/citología , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/fisiopatología , Neurotoxinas/metabolismo , Receptores Inmunológicos/genética , Receptores de Prostaglandina/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA