Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-38260577

RESUMEN

Schizophrenia (SCZ) is a genetically heterogenous psychiatric disorder of highly polygenic nature. Correlative evidence from genetic studies indicate that the aggregated effects of distinct genetic risk factor combinations found in each patient converge onto common molecular mechanisms. To prove this on a functional level, we employed a reductionistic cellular model system for polygenic risk by differentiating induced pluripotent stem cells (iPSCs) from 104 individuals with high polygenic risk load and controls into cortical glutamatergic neurons (iNs). Multi-omics profiling identified widespread differences in alternative polyadenylation (APA) in the 3' untranslated region of many synaptic transcripts between iNs from SCZ patients and healthy donors. On the cellular level, 3'APA was associated with a reduction in synaptic density of iNs. Importantly, differential APA was largely conserved between postmortem human prefrontal cortex from SCZ patients and healthy donors, and strongly enriched for transcripts related to synapse biology. 3'APA was highly correlated with SCZ polygenic risk and affected genes were significantly enriched for SCZ associated common genetic variation. Integrative functional genomic analysis identified the RNA binding protein and SCZ GWAS risk gene PTBP2 as a critical trans-acting factor mediating 3'APA of synaptic genes in SCZ subjects. Functional characterization of PTBP2 in iNs confirmed its key role in 3'APA of synaptic transcripts and regulation of synapse density. Jointly, our findings show that the aggregated effects of polygenic risk converge on 3'APA as one common molecular mechanism that underlies synaptic impairments in SCZ.

2.
Cell ; 186(23): 5165-5182.e33, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37852259

RESUMEN

Schizophrenia (SCZ) is a highly heritable mental disorder with thousands of associated genetic variants located mostly in the noncoding space of the genome. Translating these associations into insights regarding the underlying pathomechanisms has been challenging because the causal variants, their mechanisms of action, and their target genes remain largely unknown. We implemented a massively parallel variant annotation pipeline (MVAP) to perform SCZ variant-to-function mapping at scale in disease-relevant neural cell types. This approach identified 620 functional variants (1.7%) that operate in a highly developmental context and neuronal-activity-dependent manner. Multimodal integration of epigenomic and CRISPRi screening data enabled us to link these functional variants to target genes, biological processes, and ultimately alterations of neuronal physiology. These results provide a multistage prioritization strategy to map functional single-nucleotide polymorphism (SNP)-to-gene-to-endophenotype relations and offer biological insights into the context-dependent molecular processes modulated by SCZ-associated genetic variation.


Asunto(s)
Esquizofrenia , Humanos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Neuronas/metabolismo , Polimorfismo de Nucleótido Simple/genética , Esquizofrenia/genética , Animales , Ratones , Secuenciación de Nucleótidos de Alto Rendimiento
3.
bioRxiv ; 2023 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-37425902

RESUMEN

Schizophrenia (SCZ) is a highly polygenic disease and genome wide association studies have identified thousands of genetic variants that are statistically associated with this psychiatric disorder. However, our ability to translate these associations into insights on the disease mechanisms has been challenging since the causal genetic variants, their molecular function and their target genes remain largely unknown. In order to address these questions, we established a functional genomics pipeline in combination with induced pluripotent stem cell technology to functionally characterize ~35,000 non-coding genetic variants associated with schizophrenia along with their target genes. This analysis identified a set of 620 (1.7%) single nucleotide polymorphisms as functional on a molecular level in a highly cell type and condition specific fashion. These results provide a high-resolution map of functional variant-gene combinations and offer comprehensive biological insights into the developmental context and stimulation dependent molecular processes modulated by SCZ associated genetic variation.

4.
Nat Commun ; 13(1): 4819, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35974013

RESUMEN

Parkinson's disease (PD) as a progressive neurodegenerative disorder arises from multiple genetic and environmental factors. However, underlying pathological mechanisms remain poorly understood. Using multiplexed single-cell transcriptomics, we analyze human neural precursor cells (hNPCs) from sporadic PD (sPD) patients. Alterations in gene expression appear in pathways related to primary cilia (PC). Accordingly, in these hiPSC-derived hNPCs and neurons, we observe a shortening of PC. Additionally, we detect a shortening of PC in PINK1-deficient human cellular and mouse models of familial PD. Furthermore, in sPD models, the shortening of PC is accompanied by increased Sonic Hedgehog (SHH) signal transduction. Inhibition of this pathway rescues the alterations in PC morphology and mitochondrial dysfunction. Thus, increased SHH activity due to ciliary dysfunction may be required for the development of pathoetiological phenotypes observed in sPD like mitochondrial dysfunction. Inhibiting overactive SHH signaling may be a potential neuroprotective therapy for sPD.


Asunto(s)
Proteínas Hedgehog , Células-Madre Neurales , Enfermedad de Parkinson , Animales , Cilios/metabolismo , Modelos Animales de Enfermedad , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Células-Madre Neurales/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Transducción de Señal
5.
Cells ; 7(9)2018 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-30227641

RESUMEN

Schizophrenia (SCZ) is a devastating mental disorder that is characterized by distortions in thinking, perception, emotion, language, sense of self, and behavior. Epidemiological evidence suggests that subtle perturbations in early neurodevelopment increase later susceptibility for disease, which typically manifests in adolescence to early adulthood. Early perturbations are thought to be significantly mediated through incompletely understood genetic risk factors. The advent of induced pluripotent stem cell (iPSC) technology allows for the in vitro analysis of disease-relevant neuronal cell types from the early stages of human brain development. Since iPSCs capture each donor's genotype, comparison between neuronal cells derived from healthy and diseased individuals can provide important insights into the molecular and cellular basis of SCZ. In this review, we discuss results from an increasing number of iPSC-based SCZ/control studies that highlight alterations in neuronal differentiation, maturation, and neurotransmission in addition to perturbed mitochondrial function and micro-RNA expression. In light of this remarkable progress, we consider also ongoing challenges from the field of iPSC-based disease modeling that call for further improvements on the generation and design of patient-specific iPSC studies to ultimately progress from basic studies on SCZ to tailored treatments.

6.
Neuron ; 83(4): 789-96, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25123310

RESUMEN

Human induced pluripotent stem cells (iPSCs) from a healthy 86-year-old male were differentiated into neural stem cells and grafted into adult immunodeficient rats after spinal cord injury. Three months after C5 lateral hemisections, iPSCs survived and differentiated into neurons and glia and extended tens of thousands of axons from the lesion site over virtually the entire length of the rat CNS. These iPSC-derived axons extended through adult white matter of the injured spinal cord, frequently penetrating gray matter and forming synapses with rat neurons. In turn, host supraspinal motor axons penetrated human iPSC grafts and formed synapses. These findings indicate that intrinsic neuronal mechanisms readily overcome the inhibitory milieu of the adult injured spinal cord to extend many axons over very long distances; these capabilities persist even in neurons reprogrammed from very aged human cells.


Asunto(s)
Axones/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/cirugía , Anciano de 80 o más Años , Animales , Axones/ultraestructura , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Vértebras Cervicales , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/ultraestructura , Masculino , Ratas
7.
Cell Med ; 5(1): 29-42, 2013 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26858862

RESUMEN

Uniparental zygotes with two paternal (androgenetic, AG) or two maternal genomes (gynogenetic, GG) cannot develop into viable offsprings but form blastocysts from which pluripotent embryonic stem (ES) cells can be derived. For most organs, it is unclear whether uniparental ES cells can give rise to stably expandable somatic stem cells that can repair injured tissues. Even if previous reports indicated that the capacity of AG ES cells to differentiate in vitro into pan-neural progenitor cells (pNPCs) and into cells expressing neural markers is similar to biparental [normal fertilized (N)] ES cells, their potential for functional neurogenesis is not known. Here we show that murine AG pNPCs give rise to neuron-like cells, which then generate sodium-driven action potentials while maintaining fidelity of imprinted gene expression. Neural engraftment after intracerebral transplantation was achieved only by late (22 days) AG and N pNPCs with in vitro low colony-forming cell (CFC) capacity. However, persisting CFC formation seen, in particular, in early (13 or 16 days) differentiation cultures of N and AG pNPCs correlated with a high incidence of trigerm layer teratomas. As AG ES cells display functional neurogenesis and in vivo stability similar to N ES cells, they represent a unique model system to study the roles of paternal and maternal genomes on neural development and on the development of imprinting-associated brain diseases.

8.
Mol Med ; 19: 399-408, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24408113

RESUMEN

Induced cell fate changes by reprogramming of somatic cells offers an efficient strategy to generate autologous pluripotent stem (iPS) cells from any adult cell type. The potential of iPS cells to differentiate into various cell types is well established, however the efficiency to produce functional neurons from iPS cells remains modest. Here, we generated panneural progenitor cells (pNPCs) from mouse iPS cells and investigated the effect of the neurotrophic growth factor erythropoietin (EPO) on their survival, proliferation and neurodifferentiation. Under neural differentiation conditions, iPS-derived pNPCs gave rise to microtubule-associated protein-2 positive neuronlike cells (34% to 43%) and platelet-derived growth factor receptor positive oligodendrocytelike cells (21% to 25%) while less than 1% of the cells expressed the astrocytic marker glial fibrillary acidic protein. Neuronlike cells generated action potentials and developed active presynaptic terminals. The pNPCs expressed EPO receptor (EPOR) mRNA and displayed functional EPOR signaling. In proliferating cultures, EPO (0.1-3 U/mL) slightly improved pNPC survival but reduced cell proliferation and neurosphere formation in a concentration-dependent manner. In differentiating cultures EPO facilitated neurodifferentiation as assessed by the increased number of ß-III-tubulin positive neurons. Our results show that EPO inhibits iPS pNPC self-renewal and promotes neurogenesis.


Asunto(s)
Eritropoyetina/farmacología , Células Madre Pluripotentes Inducidas/fisiología , Células-Madre Neurales/fisiología , Neurogénesis , Animales , Proliferación Celular , Supervivencia Celular , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/fisiología , Oligodendroglía/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Potenciales Sinápticos
9.
PLoS One ; 7(8): e42800, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22880113

RESUMEN

Parent of origin imprints on the genome have been implicated in the regulation of neural cell type differentiation. The ability of human parthenogenetic (PG) embryonic stem cells (hpESCs) to undergo neural lineage and cell type-specific differentiation is undefined. We determined the potential of hpESCs to differentiate into various neural subtypes. Concurrently, we examined DNA methylation and expression status of imprinted genes. Under culture conditions promoting neural differentiation, hpESC-derived neural stem cells (hpNSCs) gave rise to glia and neuron-like cells that expressed subtype-specific markers and generated action potentials. Analysis of imprinting in hpESCs and in hpNSCs revealed that maternal-specific gene expression patterns and imprinting marks were generally maintained in PG cells upon differentiation. Our results demonstrate that despite the lack of a paternal genome, hpESCs generate proliferating NSCs that are capable of differentiation into physiologically functional neuron-like cells and maintain allele-specific expression of imprinted genes. Thus, hpESCs can serve as a model to study the role of maternal and paternal genomes in neural development and to better understand imprinting-associated brain diseases.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/citología , Neuronas/citología , Partenogénesis , Diferenciación Celular , Metilación de ADN/genética , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Fenómenos Electrofisiológicos/genética , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Humanos , Masculino , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/metabolismo
10.
Int J Dev Biol ; 54(11-12): 1755-62, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21404193

RESUMEN

Patient derived stem cell-based therapies are considered a future treatment option for Parkinson´s disease, a chronic and progressive brain neurodegenerative disorder characterized by depletion of dopaminergic neurons in the basal ganglia. While many aspects of the in vitro and in vivo differentiation potential of uniparental parthenogenetic (PG) and gynogenetic (GG) embryonic stem (ES) cells of several species have been studied, the capacity of androgenetic (AG) ES cells to develop into neuronal subtypes remains unclear. Here, we investigated the potential of murine AG ES cells to undergo dopaminergic differentiation both via directed in vitro differentiation, and in vivo, in ES cell-chimeric E12.5 and E16.5 brains. We show that similar to normal (N; developed from a zygote with maternal and paternal genomes) ES cells, AG cells generated dopaminergic neurons in vitro and in E12.5 and E16.5 chimeric brains following blastocyst injection. Expression of brain-specific imprinted genes was maintained in AG and normal dopaminergic cell cultures. Our results indicate that AG ES cells have dopaminergic differentiation potential in vitro and in vivo. This contrasts with previous reports of limited neural in vivo differentiation of AG cells in later brain development, and suggests that AG ES cells could be therapeutically relevant for future cellular replacement strategies for brain disease.


Asunto(s)
Encéfalo/embriología , Diferenciación Celular , Dopamina/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genoma , Neuronas/citología , Animales , Blastocisto , Encéfalo/citología , Línea Celular , Quimera/embriología , Quimera/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Células Madre , Cigoto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA