Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Cell Stem Cell ; 31(5): 676-693.e10, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38626772

RESUMO

Frontotemporal dementia (FTD) is an incurable group of early-onset dementias that can be caused by the deposition of hyperphosphorylated tau in patient brains. However, the mechanisms leading to neurodegeneration remain largely unknown. Here, we combined single-cell analyses of FTD patient brains with a stem cell culture and transplantation model of FTD. We identified disease phenotypes in FTD neurons carrying the MAPT-N279K mutation, which were related to oxidative stress, oxidative phosphorylation, and neuroinflammation with an upregulation of the inflammation-associated protein osteopontin (OPN). Human FTD neurons survived less and elicited an increased microglial response after transplantation into the mouse forebrain, which we further characterized by single nucleus RNA sequencing of microdissected grafts. Notably, downregulation of OPN in engrafted FTD neurons resulted in improved engraftment and reduced microglial infiltration, indicating an immune-modulatory role of OPN in patient neurons, which may represent a potential therapeutic target in FTD.


Assuntos
Demência Frontotemporal , Neurônios , Osteopontina , Proteínas tau , Osteopontina/metabolismo , Osteopontina/genética , Demência Frontotemporal/genética , Demência Frontotemporal/patologia , Demência Frontotemporal/metabolismo , Humanos , Neurônios/metabolismo , Neurônios/patologia , Animais , Proteínas tau/metabolismo , Camundongos , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia , Microglia/metabolismo , Microglia/patologia , Mutação/genética
2.
Mol Psychiatry ; 27(6): 2813-2820, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35365809

RESUMO

Late-onset Alzheimer's disease (LOAD) is significantly more frequent in Hispanics than in non-Hispanic Whites. Ancestry may explain these differences across ethnic groups. To this end, we studied a large cohort of Caribbean Hispanics (CH, N = 8813) and tested the association between Local Ancestry (LA) and LOAD ("admixture mapping") to identify LOAD-associated ancestral blocks, separately for ancestral components (European [EUR], African [AFR], Native American[NA]) and jointly (AFR + NA). Ancestral blocks significant after permutation were fine-mapped employing multi-ethnic whole-exome sequencing (WES) to identify rare variants associated with LOAD (SKAT-O) and replicated in the UK Biobank WES dataset. Candidate genes were validated studying (A) protein expression in human LOAD and control brains; (B) two animal AD models, Drosophila and Zebrafish. In the joint AFR + NA model, we identified four significant ancestral blocks located on chromosomes 1 (p value = 8.94E-05), 6 (p value = 8.63E-05), 21 (p value = 4.64E-05) and 22 (p value = 1.77E-05). Fine-mapping prioritized the GCAT gene on chromosome 22 (SKAT-O p value = 3.45E-05) and replicated in the UK Biobank (SKAT-O p value = 0.05). In LOAD brains, a decrease of 28% in GCAT protein expression was observed (p value = 0.038), and GCAT knockdown in Amyloid-ß42 Drosophila exacerbated rough eye phenotype (68% increase, p value = 4.84E-09). In zebrafish, gcat expression increased after acute amyloidosis (34%, p value = 0.0049), and decreased upon anti-inflammatory Interleukin-4 (39%, p value = 2.3E-05). Admixture mapping uncovered genomic regions harboring new LOAD-associated loci that might explain the observed different frequency of LOAD across ethnic groups. Our results suggest that the inflammation-related activity of GCAT is a response to amyloid toxicity, and reduced GCAT expression exacerbates AD pathology.


Assuntos
Doença de Alzheimer , Etnicidade , Doença de Alzheimer/genética , Animais , Região do Caribe , Drosophila , Humanos , Polimorfismo de Nucleotídeo Único/genética , Peixe-Zebra
3.
PLoS One ; 9(10): e110324, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25337699

RESUMO

Human motor neurons derived from embryonic and induced pluripotent stem cells (hESCs and hiPSCs) are a potentially important tool for studying motor neuron survival and pathological cell death. However, their basic survival requirements remain poorly characterized. Here, we sought to optimize a robust survival assay and characterize their response to different neurotrophic factors. First, to increase motor neuron yield, we screened a small-molecule collection and found that the Rho-associated kinase (ROCK) inhibitor Y-27632 enhances motor neuron progenitor proliferation up to 4-fold in hESC and hiPSC cultures. Next, we FACS-purified motor neurons expressing the Hb9::GFP reporter from Y-27632-amplified embryoid bodies and cultured them in the presence of mitotic inhibitors to eliminate dividing progenitors. Survival of these purified motor neurons in the absence of any other cell type was strongly dependent on neurotrophic support. GDNF, BDNF and CNTF all showed potent survival effects (EC(50) 1-2 pM). The number of surviving motor neurons was further enhanced in the presence of forskolin and IBMX, agents that increase endogenous cAMP levels. As a demonstration of the ability of the assay to detect novel neurotrophic agents, Y-27632 itself was found to support human motor neuron survival. Thus, purified human stem cell-derived motor neurons show survival requirements similar to those of primary rodent motor neurons and can be used for rigorous cell-based screening.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Fator Neurotrófico Ciliar/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , 1-Metil-3-Isobutilxantina/farmacologia , Amidas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/agonistas , AMP Cíclico/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/enzimologia , Neurônios Motores/citologia , Neurônios Motores/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
4.
Cell Rep ; 4(5): 1035-1048, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23994478

RESUMO

Differentiation of astrocytes from human stem cells has significant potential for analysis of their role in normal brain function and disease, but existing protocols generate only immature astrocytes. Using early neuralization, we generated spinal cord astrocytes from mouse or human embryonic or induced pluripotent stem cells with high efficiency. Remarkably, short exposure to fibroblast growth factor 1 (FGF1) or FGF2 was sufficient to direct these astrocytes selectively toward a mature quiescent phenotype, as judged by both marker expression and functional analysis. In contrast, tumor necrosis factor alpha and interleukin-1ß, but not FGFs, induced multiple elements of a reactive inflammatory phenotype but did not affect maturation. These phenotypically defined, scalable populations of spinal cord astrocytes will be important both for studying normal astrocyte function and for modeling human pathological processes in vitro.


Assuntos
Astrócitos/citologia , Células-Tronco Neurais/citologia , Neurônios/citologia , Células-Tronco Pluripotentes/citologia , Medula Espinal/citologia , Animais , Astrócitos/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Medula Espinal/metabolismo
5.
Biochem Pharmacol ; 86(10): 1468-75, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24021351

RESUMO

Rutin is a well-known flavonoid that exists in various natural sources. Accumulative studies have represented the biological effects of rutin, such as anti-oxidative and anti-inflammatory effects. However, the underlying mechanisms of rutin and its direct targets are not understood. We investigated whether rutin reduced B[a]PDE-induced-COX-2 expression. The transactivation of AP-1 and NF-κB were inhibited by rutin. Rutin also attenuated B[a]PDE-induced Raf/MEK/ERK and Akt activation, but had no effect on the phosphorylation of EGFR. An in vitro kinase assay revealed rutin suppressed EGFR kinase activity. We also confirmed direct binding between rutin and EGFR, and found that the binding was regressed by ATP. The EGFR inhibitor also inhibited the B[a]PDE-induced MEK/ERK and Akt signaling pathways and subsequently, suppressed COX-2 expression and promoter activity, in addition to suppressing the transactivation of AP-1 and NF-κB. In EGFR(-/-)mouse embryonic fibroblast cells, B[a]PDE-induced COX-2 expression was also diminished. Collectively, rutin inhibits B[a]PDE-induced COX-2 expression by suppressing the Raf/MEK/ERK and Akt signaling pathways. EGFR appeared to be the direct target of rutin.


Assuntos
7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/toxicidade , Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/metabolismo , Poluentes Ambientais/toxicidade , Receptores ErbB/metabolismo , Rutina/farmacologia , Animais , Linhagem Celular , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica , Quinases raf/metabolismo
6.
Rev. chil. pediatr ; 79(4): 373-380, ago. 2008. ilus
Artigo em Espanhol | LILACS | ID: lil-517480

RESUMO

Background: Few information is available about uterine effects of Cadmium (Cd) exposure, where toxic agents affecting the female genital tract interact with estrogen (E) receptors, modifiying myometrial activity and the menstrual cycle, causing dysmenorrhea, infertility and spontaneous abortion. No information exists whether prenatal or early postnatal exposure may cause any gynecologic persistent adverse effect. Our finding of a second mechanism of E interaction and differences between E receptors in the various uterine cell types suggests that Cd may affect differently E interaction in each cell-type. Objective: Evaluate a possible selective effect of acute Cd exposure on E action in the uterus during prepuber age. Method: Female prepuber rats exposed to Cd 4 mg/kg and 2 hours later, treated with Estradiol-17² 0,3 mg/kg. A myometrial sample was obtained under anaesthesia 24 hours after E treatment and histologically processed for the quantification of E responses on different uterine cell-types. Results: Cd exposure potentiates E-induced uterine eosinophilia and endometrial edema and inhibits E-induced cell hypertrophy in circular myometrium and cell proliferation in luminal myometrium. Cd, in the absence of hormone stimulation, causes a slight cell hypertrophy in circular myometrium. Conclusions: Acute exposure to Cd affects differently various responses to E in the different uterine cell-types. Future studies should verify whether this effect explains Cd-induced infertility, postpubertal sex organ development and whether prenatal or early postnatal exposure to Cd induces delayed persistent effects.


Antecedentes: Existe poca información sobre efectos del cadmio (Cd) en el útero. En mujeres altera la actividad miometrial, el ciclo menstrual y causa dismenorrea, abortos espontáneos, infertilidad y mortinatos. No existe información si la exposición prenatal o postnatal temprana causa efectos ginecológicos diferidos persistentes. Los tóxicos que afectan el útero suelen interactuar con receptores de estrógeno (E). Nuestro hallazgo de un segundo mecanismo de acción de E y de diferencias entre receptores de E de los diversos tipos celulares uterinos hacen posible que el Cd interactúe con los E en forma diferente en cada tipo celular. Objetivos: Buscar un posible efecto selectivo de la exposición aguda a Cd con algunas respuestas a E en útero de rata durante la edad prepuberal. Métodos: Ratas hembra impúberes recibieron 4 mg Cd/kg p.c. y 2 h después se trataron con 0,3 mg estradiol-17(3/kg p.c; los úteros fueron obtenidos bajo anestesia a las 24 h del tratamiento con E. Los úteros se procesaron para la cuantificación de respuestas a E en cada tipo celular por separado. Resultados: La exposición a Cd incrementa la eosinofilia uterina y edema endometrial inducidos por E; inhibe las siguientes respuestas a E: hipertrofia celular en miometrio circular, proliferación celular en epitelio luminal y miometrio. En ausencia de hormona, el cadmio causa una leve hipertrofia celular en miometrio circular. Conclusiones: La exposición aguda a Cd afecta de manera diferente las respuestas a E en los diversos tipos celulares uterinos de rata prepuberal. Futuros estudios deberán verificar si este efecto explica la infertilidad causada por exposición a Cd, afecta el desarrollo postpuberal de los órganos sexuales, e investigar si la exposición prenatal o postnatal temprana induce efectos diferidos persistentes, como puede ocurrir en población infantil prenatalmente expuesta a Cd.


Assuntos
Animais , Feminino , Cádmio/farmacologia , Cádmio/toxicidade , Doenças Uterinas/induzido quimicamente , Receptores de Estrogênio , Análise de Variância , Edema/induzido quimicamente , Endométrio , Endométrio/patologia , Eosinofilia/induzido quimicamente , Estrogênios/metabolismo , Ratos Sprague-Dawley , Útero , Útero/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA