Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 2018 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-29473171

RESUMO

Multiple sclerosis is characterised by inflammatory neurodegeneration, with axonal injury and neuronal cell death occurring in parallel to demyelination. Regarding the molecular mechanisms responsible for demyelination and axonopathy, energy failure, aberrant expression of ion channels and excitotoxicity have been suggested to lead to Ca2+ overload and subsequent activation of calcium-dependent damage pathways. Thus, the inhibition of Ca2+ influx by pharmacological modulation of Ca2+ channels may represent a novel neuroprotective strategy in the treatment of secondary axonopathy. We therefore investigated the effects of the L-type voltage-gated calcium channel blocker nimodipine in two different models of mouse experimental autoimmune encephalomyelitis (EAE), an established experimental paradigm for multiple sclerosis. We show that preventive application of nimodipine (10 mg/kg per day) starting on the day of induction had ameliorating effects on EAE in SJL/J mice immunised with encephalitic myelin peptide PLP139-151 , specifically in late-stage disease. Furthermore, supporting these data, administration of nimodipine to MOG35-55 -immunised C57BL/6 mice starting at the peak of pre-established disease, also led to a significant decrease in disease score, indicating a protective effect on secondary CNS damage. Histological analysis confirmed that nimodipine attenuated demyelination, axonal loss and pathological axonal ß-amyloid precursor protein accumulation in the cerebellum and spinal cord in the chronic phase of disease. Of note, we observed no effects of nimodipine on the peripheral immune response in EAE mice with regard to distribution, antigen-specific proliferation or activation patterns of lymphocytes. Taken together, our data suggest a CNS-specific effect of L-type voltage-gated calcium channel blockade to inflammation-induced neurodegeneration.

2.
Stem Cell Res ; 26: 76-79, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29268155

RESUMO

Human lymphoblast cells were used to generate integration-free induced pluripotent stem cells (iPSCs) employing episomal-based plasmids expressing OCT4, SOX2, NANOG, LIN28, c-MYC and L-MYC. The derived iPSCs were defined as pluripotent based on (i) expression of pluripotency-associated markers, (ii) embryoid body-based differentiation into cell types representative of the three germ layers and (iii) the similarity between the transcriptomes of the iPSC line and the human embryonic stem cell line H1 with a Pearson correlation of 0.95.


Assuntos
Diferenciação Celular , Reprogramação Celular , Corpos Embrioides/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Linfócitos/citologia , Idoso , Biomarcadores/metabolismo , Linhagem Celular , Corpos Embrioides/metabolismo , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariotipagem , Linfócitos/metabolismo
3.
Stem Cell Res ; 25: 72-82, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29112887

RESUMO

Developmental neurotoxicity (DNT) testing performed in rats is resource-intensive (costs, time, animals) and bears the issue of species extrapolation. Thus, reliable alternative human-based approaches are needed for predicting neurodevelopmental toxicity. Human induced pluripotent stem cells (hiPSCs) represent a basis for an alternative method possibly being part of an alternative DNT testing strategy. Here, we compared two hiPSC neural induction protocols resulting in 3D neurospheres: one using noggin and one cultivating cells in neural induction medium (NIM protocol). Performance of Nestin+/SOX2+ hiPSC-derived neural progenitor cells (NPCs) was compared to primary human NPCs. Generally, primary hNPCs first differentiate into Nestin+ and/or GFAP+ radial glia-like cells, while the hiPSC-derived NPCs (hiPSC-NPC) first differentiate into ßIII-Tubulin+ neurons suggesting an earlier developmental stage of hiPSC-NPC. In the 'Neurosphere Assay', NIM generated hiPSC-NPC produced neurons with higher performance than with the noggin protocol. After long-term differentiation, hiPSC-NPC form neuronal networks, which become electrically active on microelectrode arrays after 85days. Finally, methylmercury chloride inhibits hiPSC-NPC and hNPC migration with similar potencies. hiPSC-NPCs-derived neurospheres seem to be useful for DNT evaluation representing early neural development in vitro. More system characterization by compound testing is needed to gain higher confidence in this method.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Neurogênese/fisiologia , Neurônios/citologia
4.
Stem Cell Res ; 17(3): 553-555, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27789408

RESUMO

Human lymphoblast cells from a female patient diagnosed with Alzheimer's disease (AD) possessing the missense mutation TREM2 p.R47H were used to generate integration-free induced pluripotent stem cells (iPSCs) employing episomal plasmids expressing OCT4, SOX2, NANOG, LIN28, c-MYC and L-MYC. The iPSCs retained the TREM2 mutation, and were defined as pluripotent based on (i) expression of pluripotent-associated markers, (ii) embryoid body-based differentiation into cell types representative of the three germ layers and (iii) the similarity between the transcriptomes of the iPSC line and the human embryonic stem cell line H1 with a Pearson correlation of 0.961.


Assuntos
Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Glicoproteínas de Membrana/genética , Receptores Imunológicos/genética , Doença de Alzheimer/metabolismo , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Reprogramação Celular , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariótipo , Linfócitos/citologia , Microscopia de Fluorescência , Mutação de Sentido Incorreto , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos/genética , Plasmídeos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Stem Cell Res ; 17(3): 560-563, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27789410

RESUMO

Human lymphoblast cells from a female and male patient diagnosed with Alzheimer's disease (AD) with different genotypes of a functional copy number variation (CNV) in the AD risk gene CR1 were used to generate integration-free induced pluripotent stem cells (iPSCs) employing episomal plasmids expressing OCT4, SOX2, NANOG, LIN28, c-MYC and L-MYC. The iPSCs retained the CR1 CNV, and comparative transcriptome analyses with the human embryonic stem cell line H1 revealed a Pearson correlation of 0.956 for AD1-CR10 and 0.908 for AD1-CR14.


Assuntos
Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Receptores de Complemento 3b/genética , Doença de Alzheimer/genética , Diferenciação Celular , Linhagem Celular , Reprogramação Celular , Variações do Número de Cópias de DNA , Impressões Digitais de DNA , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Feminino , Dosagem de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariótipo , Linfócitos/citologia , Masculino , Microscopia de Fluorescência , Plasmídeos/genética , Plasmídeos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Oncotarget ; 7(45): 73414-73431, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27705917

RESUMO

Glioblastoma (GBM) is the most malignant brain tumor with very limited therapeutic options. Standard multimodal treatments, including surgical resection and combined radio-chemotherapy do not target the most aggressive subtype of glioma cells, brain tumor stem cells (BTSCs). BTSCs are thought to be responsible for tumor initiation, progression, and relapse. Furthermore, they have been associated with the expression of mesenchymal features as a result of epithelial-mesenchymal transition (EMT) thereby inducing tumor dissemination and chemo resistance. Using high resolution proton nuclear magnetic resonance spectroscopy (1H NMR) on GBM cell cultures we provide evidence that the expression of well-known EMT activators of the ZEB, TWIST and SNAI families and EMT target genes N-cadherin and VIMENTIN is associated with aberrant choline metabolism. The cholinic phenotype is characterized by high intracellular levels of phosphocholine and total choline derivatives and was associated with malignancy in various cancers. Both genetic and pharmacological inhibition of the cardinal choline metabolism regulator choline kinase alpha (CHKα) significantly reduces the cell viability, invasiveness, clonogenicity, and expression of EMT associated genes in GBM cells. Moreover, in some cell lines synergetic cytotoxic effects were observed when combining the standard of care chemotherapeutic temozolomide with the CHKα inhibitor V-11-0711. Taken together, specific inhibition of the enzymatic activity of CHKα is a powerful strategy to suppress EMT which opens the possibility to target chemo-resistant BTSCs through impairing their mesenchymal transdifferentiation. Moreover, the newly identified EMT-oncometabolic network may be helpful to monitor the invasive properties of glioblastomas and the success of anti-EMT therapy.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Colina/metabolismo , Transição Epitelial-Mesenquimal , Glioblastoma/metabolismo , Glioblastoma/patologia , Fenótipo , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Colina Quinase/antagonistas & inibidores , Colina Quinase/genética , Colina Quinase/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Metabolismo Energético/genética , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Técnicas de Silenciamento de Genes , Glioblastoma/genética , Humanos , Células-Tronco Neoplásicas/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Temozolomida , Vimentina/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
7.
Mol Neurodegener ; 11(1): 53, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27480121

RESUMO

BACKGROUND: Disease progression in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), as one of its animal models, is characterized by demyelination and neuronal damage in white and gray matter structures, including the hippocampus. It is thought that dysfunction of the hippocampus, a primary locus of learning and memory consolidation, may contribute to cognitive impairment in MS patients. Previously, we reported an increased generation of hippocampal neuronal progenitors in the acute stage of EAE, whereas the microenvironmental signals triggering this process remained uninvestigated. RESULTS: In the present study, we used the Wnt signaling reporter mouse Axin2(LacZ), to elucidate the molecular mechanisms underlying the activation of the hippocampal neurogenic niche upon autoimmune neuroinflammation. Histological and enzymatic examinations of ß-gal during the disease course of EAE, allowed us to survey hippocampal Wnt/ß-catenin activity, one of the key signaling pathways of adult neurogenesis. We found that Wnt signaling is transiently upregulated in the acute stage of disease, consistent with a timely induction of canonical Wnt ligands. The enhancement of signaling coincided with hippocampal neuronal damage and local expression of immune cytokines such as TNFα and IFNγ, implicating the role of the inflammatory milieu in activation of the Wnt/ß-catenin pathway. Supporting this finding, we show that transient exposure to pro-inflammatory cytokine TNFα triggers Wnt signaling in hippocampal organotypic slice cultures. Importantly, inflammation-mediated activation of the Wnt/ß-catenin pathway was associated with enhanced neurogenesis in vitro and in vivo, indicating its potential role in hippocampal tissue regeneration and repair. CONCLUSIONS: This study raises the possibility that enhancement of Wnt signaling may support neurogenic processes to cope with neuronal deficits upon immune-mediated neuroinflammation.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Hipocampo/metabolismo , Neurogênese/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Feminino , Camundongos , Esclerose Múltipla/metabolismo , beta Catenina/metabolismo
8.
Stem Cell Res ; 16(1): 29-31, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27345781

RESUMO

Human lymphoblast cells were used to generate integration-free induced pluripotent stem (iPS) cells employing episomal plasmids expressing OCT4, SOX2, NANOG, LIN28, C-MYC and L-MYC. The derived iPS cells were defined as pluripotent based on (i) expression of pluripotent-associated markers, (ii) embryoid body-based differentiation into cell types representative of the three germ layers and (iii) the similarity between the transcriptomes of the iPS cell line and the human embryonic stem cell line H1 with a Pearson correlation of 0.95.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Linfócitos/citologia , Idoso , Linhagem Celular , Humanos , Cariotipagem , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Stem Cell Res ; 16(1): 113-5, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27345793

RESUMO

Human lymphoblast cells from a male patient diagnosed with Alzheimer's disease (AD) expressing the TREM2 p.R47H variant were used to generate integration-free induced pluripotent stem (iPS) cells employing episomal plasmids expressing OCT4, SOX2, NANOG, LIN28, c-MYC and L-MYC. The iPS cells retained the TREM2 mutation, and were defined as pluripotent based on (i) expression of pluripotent-associated markers, (ii) embryoid body-based differentiation into cell types representative of the three germ layers and (iii) the similarity between the transcriptomes of the iPS cell line and the human embryonic stem cell line H1 with a Pearson correlation of 0.966.


Assuntos
Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/citologia , Glicoproteínas de Membrana/genética , Receptores Imunológicos/genética , Idoso , Doença de Alzheimer/metabolismo , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Reprogramação Celular , Análise Mutacional de DNA , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariótipo , Masculino , Glicoproteínas de Membrana/metabolismo , Plasmídeos/metabolismo , Polimorfismo de Nucleotídeo Único , Receptores Imunológicos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
10.
J Alzheimers Dis ; 50(4): 1065-82, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26890743

RESUMO

Although the incidence of Alzheimer's disease (AD) is continuously increasing in the aging population worldwide, effective therapies are not available. The interplay between causative genetic and environmental factors is partially understood. Meta-analyses have been performed on aspects such as polymorphisms, cytokines, and cognitive training. Here, we propose a meta-analysis approach based on hierarchical clustering analysis of a reliable training set of hippocampus biopsies, which is condensed to a gene expression signature. This gene expression signature was applied to various test sets of brain biopsies and iPSC-derived neuronal cell models to demonstrate its ability to distinguish AD samples from control. Thus, our identified AD-gene signature may form the basis for determination of biomarkers that are urgently needed to overcome current diagnostic shortfalls. Intriguingly, the well-described AD-related genes APP and APOE are not within the signature because their gene expression profiles show a lower correlation to the disease phenotype than genes from the signature. This is in line with the differing characteristics of the disease as early-/late-onset or with/without genetic predisposition. To investigate the gene signature's systemic role(s), signaling pathways, gene ontologies, and transcription factors were analyzed which revealed over-representation of response to stress, regulation of cellular metabolic processes, and reactive oxygen species. Additionally, our results clearly point to an important role of FOXA1 and FOXA2 gene regulatory networks in the etiology of AD. This finding is in corroboration with the recently reported major role of the dopaminergic system in the development of AD and its regulation by FOXA1 and FOXA2.


Assuntos
Doença de Alzheimer/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Hipocampo/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Transcriptoma , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Biópsia , Células Cultivadas , Redes Reguladoras de Genes , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/genética , Hipocampo/patologia , Humanos
12.
BMC Genomics ; 16: 84, 2015 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-25765079

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a complex, irreversible neurodegenerative disorder. At present there are neither reliable markers to diagnose AD at an early stage nor therapy. To investigate underlying disease mechanisms, induced pluripotent stem cells (iPSCs) allow the generation of patient-derived neuronal cells in a dish. RESULTS: In this study, employing iPS technology, we derived and characterized iPSCs from dermal fibroblasts of an 82-year-old female patient affected by sporadic AD. The AD-iPSCs were differentiated into neuronal cells, in order to generate disease-specific protein association networks modeling the molecular pathology on the transcriptome level of AD, to analyse the reflection of the disease phenotype in gene expression in AD-iPS neuronal cells, in particular in the ubiquitin-proteasome system (UPS), and to address expression of typical AD proteins. We detected the expression of p-tau and GSK3B, a physiological kinase of tau, in neuronal cells derived from AD-iPSCs. Treatment of neuronal cells differentiated from AD-iPSCs with an inhibitor of γ-secretase resulted in the down-regulation of p-tau. Transcriptome analysis of AD-iPS derived neuronal cells revealed significant changes in the expression of genes associated with AD and with the constitutive as well as the inducible subunits of the proteasome complex. The neuronal cells expressed numerous genes associated with sub-regions within the brain thus suggesting the usefulness of our in-vitro model. Moreover, an AD-related protein interaction network composed of APP and GSK3B among others could be generated using neuronal cells differentiated from two AD-iPS cell lines. CONCLUSIONS: Our study demonstrates how an iPSC-based model system could represent (i) a tool to study the underlying molecular basis of sporadic AD, (ii) a platform for drug screening and toxicology studies which might unveil novel therapeutic avenues for this debilitating neuronal disorder.


Assuntos
Doença de Alzheimer/genética , Redes Reguladoras de Genes , Neurônios/metabolismo , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Benzodiazepinonas/farmacologia , Linhagem Celular , Análise por Conglomerados , Feminino , Fibroblastos/citologia , Redes Reguladoras de Genes/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Doadores de Tecidos , Ubiquitina/genética , Ubiquitina/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
13.
Stem Cell Res Ther ; 5(1): 24, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25127410

RESUMO

The proteasome is a multi-enzyme complex responsible for orchestrating protein quality control by degrading misfolded, damaged, abnormal and foreign proteins. Studies related to the association of the proteasomal system in the preservation of self-renewal in both human and mouse pluripotent cells are sparse, and therefore a clear indication of the emergence of a new and important field of research. Under specific conditions the standard proteasome switches to the newly synthesized immunoproteasome, a catalytically active protein chamber also involved in the regulation of protein homeostasis, cell signaling and gene expression. Herein we review recent data to help elucidate and highlight the pivotal role of the proteasome complex, constitutive as well as inducible, in the regulation of self-renewal, pluripotency and differentiation of both embryonic and induced pluripotent stem cells. The proteasome that is endowed with enhanced proteolytic activity maintains self-renewal by regulating gene expression. In addition to protein degradation, the proteasome activator PA28, compartments of the 19S regulatory particle and key members of the ubiquitin pathway dictate the fate of a pluripotent stem cell. We anticipate that our observations will stimulate active research in this new and emerging theme related to stem cell biology, disease and regenerative medicine.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Humanos , Células-Tronco Pluripotentes/enzimologia , Células-Tronco Pluripotentes/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
15.
Cell ; 142(4): 613-24, 2010 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-20723761

RESUMO

Interferon (IFN)-induced immunoproteasomes (i-proteasomes) have been associated with improved processing of major histocompatibility complex (MHC) class I antigens. Here, we show that i-proteasomes function to protect cell viability under conditions of IFN-induced oxidative stress. IFNs trigger the production of reactive oxygen species, which induce protein oxidation and the formation of nascent, oxidant-damaged proteins. We find that the ubiquitylation machinery is concomitantly upregulated in response to IFNs, functioning to target defective ribosomal products (DRiPs) for degradation by i-proteasomes. i-proteasome-deficiency in cells and in murine inflammation models results in the formation of aggresome-like induced structures and increased sensitivity to apoptosis. Efficient clearance of these aggregates by the enhanced proteolytic activity of the i-proteasome is important for the preservation of cell viability upon IFN-induced oxidative stress. Our findings suggest that rather than having a specific role in the production of class I antigens, i-proteasomes increase the peptide supply for antigen presentation as part of a more general role in the maintenance of protein homeostasis.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Interferons/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas/metabolismo , Animais , Apresentação de Antígeno , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Homeostase , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ubiquitinação
16.
Nat Med ; 15(7): 788-93, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19561616

RESUMO

Previous proteomic and transcriptional analyses of multiple sclerosis lesions revealed modulation of the renin-angiotensin and the opposing kallikrein-kinin pathways. Here we identify kinin receptor B1 (Bdkrb1) as a specific modulator of immune cell entry into the central nervous system (CNS). We demonstrate that the Bdkrb1 agonist R838 (Sar-[D-Phe]des-Arg(9)-bradykinin) markedly decreases the clinical symptoms of experimental autoimmune encephalomyelitis (EAE) in SJL mice, whereas the Bdkrb1 antagonist R715 (Ac-Lys-[D-betaNal(7), Ile(8)]des-Arg(9)-bradykinin) resulted in earlier onset and greater severity of the disease. Bdkrb1-deficient (Bdkrb1(-/-)) C57BL/6 mice immunized with a myelin oligodendrocyte glycoprotein fragment, MOG(35-55), showed more severe disease with enhanced CNS-immune cell infiltration. The same held true for mixed bone marrow-chimeric mice reconstituted with Bdkrb1(-/-) T lymphocytes, which showed enhanced T helper type 17 (T(H)17) cell invasion into the CNS. Pharmacological modulation of Bdkrb1 revealed that in vitro migration of human T(H)17 lymphocytes across blood-brain barrier endothelium is regulated by this receptor. Taken together, these results suggest that the kallikrein-kinin system is involved in the regulation of CNS inflammation, limiting encephalitogenic T lymphocyte infiltration into the CNS, and provide evidence that Bdkrb1 could be a new target for the treatment of chronic inflammatory diseases such as multiple sclerosis.


Assuntos
Encéfalo/patologia , Encefalomielite Autoimune Experimental/etiologia , Receptor B1 da Bradicinina/fisiologia , Linfócitos T/fisiologia , Animais , Movimento Celular , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/terapia , Interleucina-17/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor B1 da Bradicinina/agonistas , Células Th1/fisiologia
17.
Nat Cell Biol ; 10(4): 385-94, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18344989

RESUMO

Repair processes that are activated in response to neuronal injury, be it inflammatory, ischaemic, metabolic, traumatic or other cause, are characterized by a failure to replenish neurons and by astrogliosis. The underlying molecular pathways, however, are poorly understood. Here, we show that subtle alterations of the redox state, found in different brain pathologies, regulate the fate of mouse neural progenitor cells (NPCs) through the histone deacetylase (HDAC) Sirt1. Mild oxidation or direct activation of Sirt1 suppressed proliferation of NPCs and directed their differentiation towards the astroglial lineage at the expense of the neuronal lineage, whereas reducing conditions had the opposite effect. Under oxidative conditions in vitro and in vivo, Sirt1 was upregulated in NPCs, bound to the transcription factor Hes1 and subsequently inhibited pro-neuronal Mash1. In utero shRNA-mediated knockdown of Sirt1 in NPCs prevented oxidation-mediated suppression of neurogenesis and caused upregulation of Mash1 in vivo. Our results provide evidence for an as yet unknown metabolic master switch that determines the fate of neural progenitors.


Assuntos
Diferenciação Celular/fisiologia , Neurônios/fisiologia , Sirtuínas/metabolismo , Células-Tronco/fisiologia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem da Célula , Células Cultivadas , Proteínas Correpressoras , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese/fisiologia , Neurônios/citologia , Oxirredução , Gravidez , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Sirtuína 1 , Sirtuínas/genética , Células-Tronco/citologia , Fatores de Transcrição HES-1 , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...