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










Base de dados
Intervalo de ano de publicação
1.
Restor Neurol Neurosci ; 39(4): 247-266, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34275915

RESUMO

BACKGROUND: Motor and cognitive decline as part of the normal aging process is linked to alterations in synaptic plasticity and reduction of adult neurogenesis in the dorsal striatum. Neuroinflammation, particularly in the form of microglial activation, is suggested to contribute to these age-associated changes. OBJECTIVE AND METHODS: To explore the molecular basis of alterations in striatal function during aging we analyzed RNA-Seq data for 117 postmortem human dorsal caudate samples and 97 putamen samples acquired through GTEx. RESULTS: Increased expression of neuroinflammatory transcripts including TREM2, MHC II molecules HLA-DMB, HLA-DQA2, HLA-DPA1, HLA-DPB1, HLA-DMA and HLA-DRA, complement genes C1QA, C1QB, CIQC and C3AR1, and MHCI molecules HLA-B and HLA-F was identified. We also identified down-regulation of transcripts involved in neurogenesis, synaptogenesis, and synaptic pruning, including DCX, CX3CL1, and CD200, and the canonical WNTs WNT7A, WNT7B, and WNT8A. The canonical WNT signaling pathway has previously been shown to mediate adult neurogenesis and synapse formation and growth. Recent findings also highlight the link between WNT/ß-catenin signaling and inflammation pathways. CONCLUSIONS: These findings suggest that age-dependent attenuation of canonical WNT signaling plays a pivotal role in regulating striatal plasticity during aging. Dysregulation of WNT/ß-catenin signaling via astrocyte-microglial interactions is suggested to be a novel mechanism that drives the decline of striatal neurogenesis and altered synaptic connectivity and plasticity, leading to a subsequent decrease in motor and cognitive performance with age. These findings may aid in the development of therapies targeting WNT/ß-catenin signaling to combat cognitive and motor impairments associated with aging.


Assuntos
Doenças Neuroinflamatórias , Via de Sinalização Wnt , Perfilação da Expressão Gênica , Humanos , Glicoproteínas de Membrana , Neurogênese/fisiologia , Plasticidade Neuronal/genética , Receptores Imunológicos , Via de Sinalização Wnt/genética
2.
Elife ; 82019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31794381

RESUMO

Occludin (OCLN) mutations cause human microcephaly and cortical malformation. A tight junction component thought absent in neuroepithelium after neural tube closure, OCLN isoform-specific expression extends into corticogenesis. Full-length and truncated isoforms localize to neuroprogenitor centrosomes, but full-length OCLN transiently localizes to plasma membranes while only truncated OCLN continues at centrosomes throughout neurogenesis. Mimicking human mutations, full-length OCLN depletion in mouse and in human CRISPR/Cas9-edited organoids produce early neuronal differentiation, reduced progenitor self-renewal and increased apoptosis. Human neural progenitors were more severely affected, especially outer radial glial cells, which mouse embryonic cortex lacks. Rodent and human mutant progenitors displayed reduced proliferation and prolonged M-phase. OCLN interacted with mitotic spindle regulators, NuMA and RAN, while full-length OCLN loss impaired spindle pole morphology, astral and mitotic microtubule integrity. Thus, early corticogenesis requires full-length OCLN to regulate centrosome organization and dynamics, revealing a novel role for this tight junction protein in early brain development.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Ocludina/metabolismo , Junções Íntimas/metabolismo , Aneuploidia , Animais , Apoptose , Sistemas CRISPR-Cas , Diferenciação Celular , Proliferação de Células , Centrossomo/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Edição de Genes , Humanos , Camundongos , Camundongos Knockout , Microcefalia/genética , Microcefalia/patologia , Microtúbulos/metabolismo , Mutagênese , Mutação , Neurogênese/genética , Neurogênese/fisiologia , Ocludina/genética , Fuso Acromático/metabolismo , Junções Íntimas/genética
3.
Neurobiol Stress ; 10: 100145, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30937351

RESUMO

Prenatal substance exposure is a growing public health concern worldwide. Although the opioid crisis remains one of the most prevalent addiction problems in our society, abuse of cocaine, methamphetamines, and other illicit drugs, particularly amongst pregnant women, are nonetheless significant and widespread. Evidence demonstrates prenatal drug exposure can affect fetal brain development and thus can have long-lasting impact on neurobehavioral and cognitive performance later in life. In this review, we highlight research examining the most prevalent drugs of abuse and their effects on brain development with a focus on endoplasmic reticulum stress and oxidative stress signaling pathways. A thorough exploration of drug-induced cellular stress mechanisms during prenatal brain development may provide insight into therapeutic interventions to combat effects of prenatal drug exposure.

4.
Neuron ; 95(3): 482-485, 2017 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-28772118

RESUMO

Brain function requires connecting neuronal networks to empower movement, sensation, behavior, and cognition. Studies published early this year provide evidence that in humans, Netrin receptor, Deleted in Colorectal Cancer (DCC), is a master regulator of axonal crossing throughout the neuraxis.


Assuntos
Axônios/fisiologia , Encéfalo/metabolismo , Cones de Crescimento/fisiologia , Fatores de Crescimento Neural/metabolismo , Neurônios/fisiologia , Receptores de Superfície Celular/metabolismo , Animais , Humanos , Receptores de Netrina
5.
J Biomed Sci ; 24(1): 59, 2017 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-28822354

RESUMO

Three-dimensional (3D) brain organoids derived from human pluripotent stem cells (hPSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), appear to recapitulate the brain's 3D cytoarchitectural arrangement and provide new opportunities to explore disease pathogenesis in the human brain. Human iPSC (hiPSC) reprogramming methods, combined with 3D brain organoid tools, may allow patient-derived organoids to serve as a preclinical platform to bridge the translational gap between animal models and human clinical trials. Studies using patient-derived brain organoids have already revealed novel insights into molecular and genetic mechanisms of certain complex human neurological disorders such as microcephaly, autism, and Alzheimer's disease. Furthermore, the combination of hiPSC technology and small-molecule high-throughput screening (HTS) facilitates the development of novel pharmacotherapeutic strategies, while transcriptome sequencing enables the transcriptional profiling of patient-derived brain organoids. Finally, the addition of CRISPR/Cas9 genome editing provides incredible potential for personalized cell replacement therapy with genetically corrected hiPSCs. This review describes the history and current state of 3D brain organoid differentiation strategies, a survey of applications of organoids towards studies of neurodevelopmental and neurodegenerative disorders, and the challenges associated with their use as in vitro models of neurological disorders.


Assuntos
Encéfalo/patologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Doenças Neurodegenerativas/patologia , Transtornos do Neurodesenvolvimento/patologia , Organoides/fisiologia , Animais , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular , Modelos Animais de Doenças , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia
6.
Psychiatry Clin Neurosci ; 71(10): 663-672, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28675555

RESUMO

RNA sequencing (RNA-Seq), a revolutionary tool for transcriptome profiling, is becoming increasingly important for neuroscientists in studying the transcriptional landscape of the human brain. Studies using this next-generation sequencing technique have already revealed novel insights into the complexity of neurons in the human brain and pathogenesis of complex neurological diseases. In clinical neuroscience, RNA-Seq provides exciting opportunities for improving diagnosis and treatment of neurological diseases by facilitating the development of pharmacotherapies able to modulate gene expression. Furthermore, integrative whole genome sequencing and transcriptome sequencing can provide additional information for the functional role of mutated genes, prioritization of variants, and intron/exon splicing. This review describes the current state of RNA-Seq studies in neuropsychiatric disorders using post-mortem human brains, a brief survey of best practices for experimental design and sequencing data analysis, and the challenges associated with its application in the human brain.


Assuntos
Encéfalo/metabolismo , Transtornos Mentais/genética , Análise de Sequência de RNA/métodos , Perfilação da Expressão Gênica/métodos , Humanos , Projetos de Pesquisa , Estatística como Assunto/métodos , Sequenciamento Completo do Genoma/métodos
7.
Neuropsychopharmacology ; 42(3): 774-784, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27534267

RESUMO

Because of unavoidable confounding variables in the direct study of human subjects, it has been difficult to unravel the effects of prenatal cocaine exposure on the human fetal brain, as well as the cellular and biochemical mechanisms involved. Here, we propose a novel approach using a human pluripotent stem cell (hPSC)-based 3D neocortical organoid model. This model retains essential features of human neocortical development by encompassing a single self-organized neocortical structure, without including an animal-derived gelatinous matrix. We reported previously that prenatal cocaine exposure to rats during the most active period of neural progenitor proliferation induces cytoarchitectural changes in the embryonic neocortex. We also identified a role of CYP450 and consequent oxidative ER stress signaling in these effects. However, because of differences between humans and rodents in neocorticogenesis and brain CYP metabolism, translation of the research findings from the rodent model to human brain development is uncertain. Using hPSC 3D neocortical organoids, we demonstrate that the effects of cocaine are mediated through CYP3A5-induced generation of reactive oxygen species, inhibition of neocortical progenitor cell proliferation, induction of premature neuronal differentiation, and interruption of neural tissue development. Furthermore, knockdown of CYP3A5 reversed these cocaine-induced pathological phenotypes, suggesting CYP3A5 as a therapeutic target to mitigate the deleterious neurodevelopmental effects of prenatal cocaine exposure in humans. Moreover, 3D organoid methodology provides an innovative platform for identifying adverse effects of abused psychostimulants and pharmaceutical agents, and can be adapted for use in neurodevelopmental disorders with genetic etiologies.


Assuntos
Cocaína/farmacologia , Citocromo P-450 CYP3A/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Neocórtex/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Linhagem Celular , Humanos
8.
Restor Neurol Neurosci ; 33(3): 347-56, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25835555

RESUMO

PURPOSE: We describe a technique for independently differentiating neocortical and mesencephalic dopaminergic (mDA) neurons from a single human pluripotent stem cell (hPSC) line, and subsequently allowing the two cell types to interact and form connections. METHODS: Dopaminergic and neocortical progenitors were differentiated in separate vessels, then separately seeded into the inner and outer compartments of specialized cell culture vessels designed for in vitro studies of wound healing. Cells were further differentiated using dopamine-specific and neocortex-specific trophic factors, respectively. The barrier was then removed, and differentiation was continued for three weeks in the presence of BDNF. RESULTS: After three weeks of differentiation, neocortical and mDA cell bodies largely remained in the areas into which they had been seeded, and the gap between the mDA and neocortical neuron populations could still be discerned. Abundant tyrosine hydroxylase (TH)-positive projections had extended from the area of the inner chamber to the outer chamber neocortical area. CONCLUSIONS: We have developed a hPSC-based system for producing connections between neurons from two brain regions, neocortex and midbrain. Future experiments could employ modifications of this method to examine connections between any two brain regions or neuronal subtypes that can be produced from hPSCs in vitro.


Assuntos
Comunicação Celular/fisiologia , Neurônios Dopaminérgicos/citologia , Mesencéfalo/citologia , Neurogênese/fisiologia , Células-Tronco Pluripotentes/citologia , Humanos
9.
Cell Rep ; 10(4): 616-32, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25640183

RESUMO

Human pluripotent stem cell (hPSC) lines exhibit repeated patterns of genetic variation, which can alter in vitro properties as well as suitability for clinical use. We examined associations between copy-number variations (CNVs) on chromosome 17 and hPSC mesodiencephalic dopaminergic (mDA) differentiation. Among 24 hPSC lines, two karyotypically normal lines, BG03 and CT3, and BG01V2, with trisomy 17, exhibited amplification of the WNT3/WNT9B region and rapid mDA differentiation. In hPSC lines with amplified WNT3/WNT9B, basic fibroblast growth factor (bFGF) signaling through mitogen-activated protein kinase (MAPK)/ERK amplifies canonical WNT signaling by phosphorylating LRP6, resulting in enhanced undifferentiated proliferation. When bFGF is absent, noncanonical WNT signaling becomes dominant due to upregulation of SIAH2, enhancing JNK signaling and promoting loss of pluripotency. When bFGF is present during mDA differentiation, stabilization of canonical WNT signaling causes upregulation of LMX1A and mDA induction. Therefore, CNVs in 17q21.31, a "hot spot" for genetic variation, have multiple and complex effects on hPSC cellular phenotype.


Assuntos
Neurônios/citologia , Neurônios/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt3/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Proteínas Wnt/genética , Proteína Wnt3/genética
10.
Dis Model Mech ; 7(12): 1397-405, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25288682

RESUMO

Neocortical development involves ordered specification of forebrain cortical progenitors to various neuronal subtypes, ultimately forming the layered cortical structure. Modeling of this process using human pluripotent stem cells (hPSCs) would enable mechanistic studies of human neocortical development, while providing new avenues for exploration of developmental neocortical abnormalities. Here, we show that preserving hPSCs aggregates - allowing embryoid body formation - while adding basic fibroblast growth factor (bFGF) during neuroepithelial development generates neural rosettes showing dorsal forebrain identity, including Mash1(+) dorsal telencephalic GABAergic progenitors. Structures that mirrored the organization of the cerebral cortex formed after rosettes were seeded and cultured for 3 weeks in the presence of FGF18, BDNF and NT3. Neurons migrated along radial glia scaffolding, with deep-layer CTIP2(+) cortical neurons appearing after 1 week and upper-layer SATB2(+) cortical neurons forming during the second and third weeks. At the end of differentiation, these structures contained both glutamatergic and GABAergic neurons, with glutamatergic neurons being most abundant. Thus, this differentiation protocol generated an hPSC-based model that exhibits temporal patterning and a neuronal subtype ratio similar to that of the developing human neocortex. This model was used to examine the effects of cocaine during neocorticogenesis. Cocaine caused premature neuronal differentiation and enhanced neurogenesis of various cortical neuronal subtypes. These cocaine-induced changes were inhibited by the cytochrome P450 inhibitor cimetidine. This in vitro model enables mechanistic studies of neocorticogenesis, and can be used to examine the mechanisms through which cocaine alters the development of the human neocortex.


Assuntos
Córtex Cerebral/embriologia , Cocaína/química , Células-Tronco Pluripotentes/citologia , Diferenciação Celular , Linhagem Celular , Inibidores da Captação de Dopamina/química , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica , Neurogênese , Neurônios/metabolismo , Prosencéfalo/embriologia , Espécies Reativas de Oxigênio , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...