Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Glia ; 70(1): 173-195, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34661306

RESUMO

Microglia cells are active players in regulating synaptic development and plasticity in the brain. However, how they influence the normal functioning of synapses is largely unknown. In this study, we characterized the effects of pharmacological microglia depletion, achieved by administration of PLX5622, on hippocampal CA3-CA1 synapses of adult wild type mice. Following microglial depletion, we observed a reduction of spontaneous and evoked glutamatergic activity associated with a decrease of dendritic spine density. We also observed the appearance of immature synaptic features and higher levels of plasticity. Microglia depleted mice showed a deficit in the acquisition of the Novel Object Recognition task. These events were accompanied by hippocampal astrogliosis, although in the absence ofneuroinflammatory condition. PLX-induced synaptic changes were absent in Cx3cr1-/- mice, highlighting the role of CX3CL1/CX3CR1 axis in microglia control of synaptic functioning. Remarkably, microglia repopulation after PLX5622 withdrawal was associated with the recovery of hippocampal synapses and learning functions. Altogether, these data demonstrate that microglia contribute to normal synaptic functioning in the adult brain and that their removal induces reversible changes in organization and activity of glutamatergic synapses.


Assuntos
Microglia , Neurônios , Animais , Encéfalo , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Hipocampo , Camundongos , Compostos Orgânicos/farmacologia , Sinapses/fisiologia
2.
Gut ; 69(11): 2016-2024, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32114505

RESUMO

OBJECTIVE: The HBV HBx regulatory protein is required for transcription from the covalently closed circular DNA (cccDNA) minichromosome and affects the epigenetic control of both viral and host cellular chromatin. DESIGN: We explored, in relevant cellular models of HBV replication, the functional consequences of HBx interaction with DLEU2, a long non-coding RNA (lncRNA) expressed in the liver and increased in human hepatocellular carcinoma (HCC), in the regulation of host target genes and the HBV cccDNA. RESULTS: We show that HBx binds the promoter region, enhances the transcription and induces the accumulation of DLEU2 in infected hepatocytes. We found that nuclear DLEU2 directly binds HBx and the histone methyltransferase enhancer of zeste homolog 2 (EZH2), the catalytic active subunit of the polycomb repressor complex 2 (PRC2) complex. Computational modelling and biochemical evidence suggest that HBx and EZH2 share two preferential binding sites in DLEU2 intron 1. HBx and DLEU2 co-recruitment on the cccDNA displaces EZH2 from the viral chromatin to boost transcription and viral replication. DLEU2-HBx association with target host promoters relieves EZH2 repression and leads to the transcriptional activation of a subset of EZH2/PRC2 target genes in HBV-infected cells and HBV-related HCCs. CONCLUSIONS: Our results highlight the ability of HBx to bind RNA to impact on the epigenetic control of both viral cccDNA and host genes and provide a new key to understand the role of DLEU2 and EZH2 overexpression in HBV-related HCCs and HBx contribution to hepatocytes transformation.


Assuntos
Carcinoma Hepatocelular/etiologia , Vírus da Hepatite B/fisiologia , Hepatócitos/patologia , Neoplasias Hepáticas/etiologia , Transativadores/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Replicação Viral/fisiologia , Técnicas de Cultura de Células , DNA Circular , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Hepatócitos/metabolismo , Humanos , RNA Longo não Codificante/metabolismo
3.
Biochem Biophys Res Commun ; 461(1): 42-6, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25858320

RESUMO

The Sir proteins, namely Sir2, 3 and 4, have roles related to heterochromatin, but genome-wide studies have revealed their presence at many euchromatic loci, although the functional meaning of this is still not clear. Nhp6a is an abundant HMG-like protein in yeast, which has a role in transcription by modulating chromatin structure and nucleosome number. Although much is known about its structure and function, information regarding its regulation is scarce. NHP6A, among other genes, emerges in ChIP-on chip studies of global Sir proteins binding, suggesting it could be regulated by SIR. We have investigated NHP6A expression in sir deletion mutants as well as in SIR2 overexpressing conditions. In addition, we have asked if the Sir2 deacetylation activity is involved by using conditions that either inhibit (treatment with nicotinamide) or enhance (calorie restriction conditions) Sir2 activity. We have found that, consistent with previous microarray studies, NHP6A expression undergoes a slight increase in sir mutant strains, but is strongly repressed when SIR2 is overexpressed. In a sir3 mutant strain the gene continues to be transcribed, even in SIR2 overexpressing conditions. In addition, treating the cells with nicotinamide counteracts the SIR2 overexpressing effect. Finally, conditions that are known to potentiate Sir2 deacetylation activity seem to mimic the effect of SIR2 overexpression on NHP6A. Our results suggest that Sir2 is involved in the regulation of NHP6A promoter, acting more as a specific repressor, rather than a long-range silencer. This effect is specific, and the Sir2 deacetylase activity is required for the Sir2 mediated repression of NHP6A. Moreover, the presence of the SIR complex seems required for Sir2 to silence NHP6A.


Assuntos
Regulação Fúngica da Expressão Gênica/genética , Proteínas HMGN/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Informação Silenciosa de Saccharomyces cerevisiae/genética , Sirtuína 2/genética , Ativação Transcricional/genética , Restrição Calórica , Transdução de Sinais/genética , Estresse Fisiológico/genética
4.
Gene ; 913: 148387, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38499211

RESUMO

BACKGROUND: Epigenetic mechanisms, including DNA methylation, histone modifications, and chromatin remodeling, are highly involved in the regulation of hepatocyte viability, proliferation, and plasticity. We have previously demonstrated that repression of H3K27 methylation in differentiated hepatic HepaRG cells by treatment with GSK-J4, an inhibitor of JMJD3 and UTX H3K27 demethylase activity, changed their phenotype, inducing differentiated hepatocytes to proliferate. In addition to the epigenetic enzymatic role in the regulation of the retro-differentiation process, emerging evidence indicate that microRNAs (miRNAs) are involved in controlling hepatocyte proliferation during liver regeneration. Hence, the aim of this work is to investigate the impact of H3K27 methylation on miRNAs expression profile and its role in the regulation of the differentiation status of human hepatic progenitors HepaRG cells. METHODS: A miRNA-sequencing was carried out in differentiated HepaRG cells treated or not with GSK-J4. Target searching and Gene Ontology analysis were performed to identify the molecular processes modulated by differentially expressed miRNAs. The biological functions of selected miRNAs was further investigated by transfection of miRNAs inhibitors or mimics in differentiated HepaRG cells followed by qPCR analysis, albumin ELISA assay, CD49a FACS analysis and EdU staining. RESULTS: We identified 12 miRNAs modulated by GSK-J4; among these, miR-27a-3p and miR- 423-5p influenced the expression of several proliferation genes in differentiated HepaRG cells. MiR-27a-3p overexpression increased the number of hepatic cells reentering proliferation. Interestingly, both miR-27a-3p and miR-423-5p did not affect the expression levels of genes involved in the differentiation of progenitors HepaRG cells. CONCLUSIONS: Modulation of H3K27me3 methylation in differentiated HepaRG cells, by GSK-J4 treatment, influenced miRNA' s expression profile pushing liver cells towards a proliferating phenotype. We demonstrated the involvement of miR-27a-3p in reinducing proliferation of differentiated hepatocytes suggesting a potential role in liver plasticity.


Assuntos
Hepatócitos , MicroRNAs , Humanos , Hepatócitos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fígado/metabolismo , Diferenciação Celular/genética , Epigênese Genética
5.
Cell Death Discov ; 9(1): 33, 2023 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36702823

RESUMO

Mutations in RNA binding proteins (RBPs) have been linked to the motor neuron disease amyotrophic lateral sclerosis (ALS). Extensive auto-regulation, cross-regulation, cooperation and competition mechanisms among RBPs are in place to ensure proper expression levels of common targets, often including other RBPs and their own transcripts. Moreover, several RBPs play a crucial role in the nervous system by localizing target RNAs in specific neuronal compartments. These include the RBPs FUS, FMRP, and HuD. ALS mutations in a given RBP are predicted to produce a broad impact on such delicate equilibrium. Here we studied the effects of the severe FUS-P525L mutation on common FUS and FMRP targets. Expression profiling by digital color-coded molecular barcoding in cell bodies and neurites of human iPSC-derived motor neurons revealed altered levels of transcripts involved in the cytoskeleton, neural projection and synapses. One of the common targets is HuD, which is upregulated because of the loss of FMRP binding to its 3'UTR due to mutant FUS competition. Notably, many genes are commonly altered upon FUS mutation or HuD overexpression, suggesting that a substantial part of the effects of mutant FUS on the motor neuron transcriptome could be due to HuD gain-of-function. Among altered transcripts, we also identified other common FUS and FMRP targets, namely MAP1B, PTEN, and AP2B1, that are upregulated upon loss of FMRP binding on their 3'UTR in FUS-P525L motor neurons. This work demonstrates that the impairment of FMRP function by mutant FUS might alter the expression of several genes, including new possible biomarkers and therapeutic targets for ALS.

6.
Stem Cells Int ; 2020: 8852313, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33005195

RESUMO

Human pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells, represent powerful tools for disease modeling and for therapeutic applications. PSCs are particularly useful for the study of development and diseases of the nervous system. However, generating in vitro models that recapitulate the architecture and the full variety of subtypes of cells that make the complexity of our brain remains a challenge. In order to fully exploit the potential of PSCs, advanced methods that facilitate the identification of molecular signatures in neural differentiation and neurological diseases are highly demanded. Here, we review the literature on the development and application of digital color-coded molecular barcoding as a potential tool for standardizing PSC research and applications in neuroscience. We will also describe relevant examples of the use of this technique for the characterization of the heterogeneous composition of the brain tumor glioblastoma multiforme.

7.
Cell Death Dis ; 10(7): 518, 2019 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-31285428

RESUMO

Modification of histones by lysine methylation plays a role in many biological processes, and it is dynamically regulated by several histone methyltransferases and demethylases. The polycomb repressive complex contains the H3K27 methyltransferase EZH2 and controls dimethylation and trimethylation of H3K27 (H3K27me2/3), which trigger gene suppression. JMJD3 and UTX have been identified as H3K27 demethylases that catalyze the demethylation of H3K27me2/3, which in turns lead to gene transcriptional activation. EZH2, JMJD3 and UTX have been extensively studied for their involvement in development, immune system, neurodegenerative disease, and cancer. However, their role in molecular mechanisms underlying the differentiation process of hepatic cells is yet to be elucidated. Here, we show that EZH2 methyltransferase and JMJD3/UTX demethylases were deregulated during hepatic differentiation of human HepaRG cells resulting in a strong reduction of H3K27 methylation levels. Inhibition of JMJD3 and UTX H3K27 demethylase activity by GSK-J4 epi-drug reverted phenotype of HepaRG DMSO-differentiated cells and human primary hepatocytes, drastically decreasing expression of hepatic markers and inducing cell proliferation. In parallel, inhibition of EZH2 H3K27me3 activity by GSK-126 epi-drug induced upregulation of hepatic markers and downregulated the expression of cell cycle inhibitor genes. To conclude, we demonstrated that modulation of H3K27 methylation by inhibiting methyl-transferase and dimethyl-transferase activity influences the differentiation status of hepatic cells, identifying a possible new role of EZH2, JMJD3 and UTX epi-drugs to modulate hepatic cell plasticity.


Assuntos
Diferenciação Celular , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Histona Desmetilases/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Fígado/citologia , Benzazepinas/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Epigênese Genética/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Histonas/metabolismo , Humanos , Indóis/farmacologia , Lisina/metabolismo , Metilação , Análise de Componente Principal , Piridonas/farmacologia , Pirimidinas/farmacologia
8.
J Vis Exp ; (149)2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31380833

RESUMO

Hepatic steatosis represents a metabolic dysfunction that results from an accumulation of triglyceride-containing lipid droplets in hepatocytes. Excessive fat accumulation leads to non-alcoholic fatty liver disease (NAFLD),  which is potentially reversible and may evolve into non-alcoholic steatohepatitis (NASH) and eventually cirrhosis and hepatocellular carcinoma (HCC). The molecular mechanisms linking lipid accumulation in hepatocytes with the progression to NASH, irreversible liver damage, fibrosis, cirrhosis, and even HCC still remains unclear. To this end, several in vitro and in vivo models have been developed to elucidate the pathological processes that cause NAFLD. In the present study, we describe a cellular model for the induction of liver vesicular steatosis that consists of DMSO-differentiated human hepatic HepaRG cells treated with the fatty acid salt sodium oleate. Indeed, sodium oleate-treated HepaRG cells accumulate lipid droplets in the cytoplasm and show typical features of steatosis. This in vitro human model represents a valuable alternative to in vivo mice models as well as to the primary human hepatocytes. We also present a comparison of several methods for the quantification and evaluation of fat accumulation in HepaRG cells, including Oil Red O staining, cytofluorimetric Bodipy measurement, metabolic gene expression analysis by qPCR, and coherent anti-Stokes Raman scattering (CARS) microscopy. CARS imaging combines the chemical specificity of Raman spectroscopy, a chemical analysis technique well-known in materials science applications, with the benefits of high-speed, high-resolution non-linear optical microscopies to allow precise quantification of lipid accumulation and lipid droplet dynamics. The establishment of an efficient in vitro model for the induction of vesicular steatosis, alongside an accurate method for the quantification and characterization of lipid accumulation, could lead to the development of early stage diagnosis of NAFLD via the identification of molecular markers, and to the generation of new treatment strategies.


Assuntos
Diferenciação Celular , Fígado Gorduroso/patologia , Hepatócitos/patologia , Animais , Linhagem Celular , Fígado Gorduroso/metabolismo , Hepatócitos/metabolismo , Humanos , Camundongos
9.
PLoS One ; 13(1): e0189477, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29320502

RESUMO

Individuals exposed to Mycobacterium tuberculosis (Mtb) may be infected and remain for the entire life in this condition defined as latent tuberculosis infection (LTBI) or develop active tuberculosis (TB). Among the multiple factors governing the outcome of the infection, dendritic cells (DCs) play a major role in dictating antibacterial immunity. However, current knowledge on the role of the diverse components of human DCs in shaping specific T-cell response during Mtb infection is limited. In this study, we performed a comparative evaluation of peripheral blood circulating DC subsets as well as of monocyte-derived Interferon-α DCs (IFN-DCs) from patients with active TB, subjects with LTBI and healthy donors (HD). The proportion of circulating myeloid BDCA3+ DCs (mDC2) and plasmacytoid CD123+ DCs (pDCs) declined significantly in active TB patients compared to HD, whereas the same subsets displayed a remarkable activation in LTBI subjects. Simultaneously, the differentiation of IFN-DCs from active TB patients resulted profoundly impaired compared to those from LTBI and HD individuals. Importantly, the altered developmental trait of IFN-DCs from active TB patients was associated with down-modulation of IFN-linked genes, marked changes in molecular signaling conveying antigen (Ag) presentation and full inability to induce Ag-specific T cell response. Thus, these data reveal an important role of IFN-α in determining the induction of Mtb-specific immunity.


Assuntos
Células Dendríticas/imunologia , Interferon-alfa/imunologia , Tuberculose Latente/patologia , Adulto , Antígenos CD/imunologia , Regulação para Baixo , Feminino , Humanos , Tuberculose Latente/imunologia , Masculino , Pessoa de Meia-Idade , Mycobacterium tuberculosis/imunologia
10.
Sci Rep ; 8(1): 13638, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30206377

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver disease. Although genetic predisposition and epigenetic factors contribute to the development of NAFLD, our understanding of the molecular mechanism involved in the pathogenesis of the disease is still emerging. Here we investigated a possible role of a microRNAs-STAT3 pathway in the induction of hepatic steatosis. Differentiated HepaRG cells treated with the fatty acid sodium oleate (fatty dHepaRG) recapitulated features of liver vesicular steatosis and activated a cell-autonomous inflammatory response, inducing STAT3-Tyrosine-phosphorylation. With a genome-wide approach (Chromatin Immunoprecipitation Sequencing), many phospho-STAT3 binding sites were identified in fatty dHepaRG cells and several STAT3 and/or NAFLD-regulated microRNAs showed increased expression levels, including miR-21. Innovative CARS (Coherent Anti-Stokes Raman Scattering) microscopy revealed that chemical inhibition of STAT3 activity decreased lipid accumulation and deregulated STAT3-responsive microRNAs, including miR-21, in lipid overloaded dHepaRG cells. We were able to show in vivo that reducing phospho-STAT3-miR-21 levels in C57/BL6 mice liver, by long-term treatment with metformin, protected mice from aging-dependent hepatic vesicular steatosis. Our results identified a microRNAs-phosphoSTAT3 pathway involved in the development of hepatic steatosis, which may represent a molecular marker for both diagnosis and therapeutic targeting.


Assuntos
Envelhecimento/metabolismo , Fígado Gorduroso/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Metformina/farmacologia , MicroRNAs/metabolismo , Fator de Transcrição STAT3/metabolismo , Envelhecimento/patologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/patologia , Estudo de Associação Genômica Ampla , Camundongos , Microscopia Óptica não Linear , Fosforilação/efeitos dos fármacos
11.
Nat Med ; 23(3): 327-336, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28165481

RESUMO

Hepatitis B virus (HBV)-specific CD8 T cells are functionally exhausted in chronic hepatitis B infection, and this condition can be corrected only partially through the modulation of inhibitory pathways, which suggests that a more complex molecular interplay underlies T cell exhaustion. To gain broader insight into this process and identify additional targets for the restoration of T cell function, we compared the transcriptome profiles of HBV-specific CD8 T cells from patients with acute and chronic disease with those of HBV-specific CD8 T cells from patients able to resolve HBV infection spontaneously and influenza (FLU)-specific CD8 T cells from healthy participants. The results indicate that exhausted HBV-specific CD8 T cells are markedly impaired at multiple levels and show substantial downregulation of various cellular processes centered on extensive mitochondrial alterations. A notable improvement of mitochondrial and antiviral CD8 functions was elicited by mitochondrion-targeted antioxidants, which suggests a central role for reactive oxygen species (ROS) in T cell exhaustion. Thus, mitochondria represent promising targets for novel reconstitution therapies to treat chronic hepatitis B infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Hepatite B Crônica/imunologia , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Doença Aguda , Adulto , Idoso , Antioxidantes/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Citocinas/imunologia , Regulação para Baixo , Feminino , Hepatite B/imunologia , Hepatite B Crônica/genética , Hepatite B Crônica/metabolismo , Humanos , Masculino , Potencial da Membrana Mitocondrial , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Reação em Cadeia da Polimerase , Superóxidos/metabolismo , Transcriptoma , Adulto Jovem
12.
Sci Rep ; 7(1): 1093, 2017 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-28439087

RESUMO

Immunotherapy efficacy relies on the crosstalk within the tumor microenvironment between cancer and dendritic cells (DCs) resulting in the induction of a potent and effective antitumor response. DCs have the specific role of recognizing cancer cells, taking up tumor antigens (Ags) and then migrating to lymph nodes for Ag (cross)-presentation to naïve T cells. Interferon-α-conditioned DCs (IFN-DCs) exhibit marked phagocytic activity and the special ability of inducing Ag-specific T-cell response. Here, we have developed a novel microfluidic platform recreating tightly interconnected cancer and immune systems with specific 3D environmental properties, for tracking human DC behaviour toward tumor cells. By combining our microfluidic platform with advanced microscopy and a revised cell tracking analysis algorithm, it was possible to evaluate the guided efficient motion of IFN-DCs toward drug-treated cancer cells and the succeeding phagocytosis events. Overall, this platform allowed the dissection of IFN-DC-cancer cell interactions within 3D tumor spaces, with the discovery of major underlying factors such as CXCR4 involvement and underscored its potential as an innovative tool to assess the efficacy of immunotherapeutic approaches.


Assuntos
Rastreamento de Células/métodos , Neoplasias do Colo/terapia , Células Dendríticas/imunologia , Imunoterapia/métodos , Microfluídica/métodos , Modelos Biológicos , Células Cultivadas , Humanos , Fatores Imunológicos/metabolismo , Interferon-alfa/metabolismo , Microscopia/métodos , Resultado do Tratamento
13.
PLoS One ; 10(11): e0142599, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26580974

RESUMO

The HBV covalently closed circular DNA (cccDNA) is organized as a mini-chromosome in the nuclei of infected hepatocytes by histone and non-histone proteins. Transcription from the cccDNA of the RNA replicative intermediate termed pre-genome (pgRNA), is the critical step for genome amplification and ultimately determines the rate of HBV replication. Multiple evidences suggest that cccDNA epigenetic modifications, such as histone modifications and DNA methylation, participate in regulating the transcriptional activity of the HBV cccDNA. Inflammatory cytokines (TNFα, LTß) and the pleiotropic cytokine interleukin-6 (IL6) inhibit hepatitis B virus (HBV) replication and transcription. Here we show, in HepG2 cells transfected with linear HBV monomers and HBV-infected NTCP-HepG2 cells, that IL6 treatment leads to a reduction of cccDNA-bound histone acetylation paralleled by a rapid decrease in 3.5kb/pgRNA and subgenomic HBV RNAs transcription without affecting cccDNA chromatinization or cccDNA levels. IL6 repressive effect on HBV replication is mediated by a loss of HNF1α and HNF4α binding to the cccDNA and a redistribution of STAT3 binding from the cccDNA to IL6 cellular target genes.


Assuntos
Vírus da Hepatite B/genética , Hepatite B Crônica/genética , Interleucina-6/genética , Transcrição Gênica , Replicação Viral/genética , Metilação de DNA/genética , DNA Circular/genética , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Epigênese Genética , Regulação Viral da Expressão Gênica , Células Hep G2 , Vírus da Hepatite B/patogenicidade , Hepatite B Crônica/virologia , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/genética , Histonas/genética , Humanos , Interleucina-6/metabolismo , RNA/genética , Fator de Transcrição STAT3/genética
14.
PLoS One ; 8(8): e72833, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977359

RESUMO

Type I interferon (IFN-I) have emerged as crucial mediators of cellular signals controlling DC differentiation and function. Human DC differentiated from monocytes in the presence of IFN-α (IFN-α DC) show a partially mature phenotype and a special capability of stimulating CD4+ T cell and cross-priming CD8+ T cells. Likewise, plasmacytoid DC (pDC) are blood DC highly specialized in the production of IFN-α in response to viruses and other danger signals, whose functional features may be shaped by IFN-I. Here, we investigated the molecular mechanisms stimulated by IFN-α in driving human monocyte-derived DC differentiation and performed parallel studies on peripheral unstimulated and IFN-α-treated pDC. A specific miRNA signature was induced in IFN-α DC and selected miRNAs, among which miR-23a and miR-125b, proved to be negatively associated with up-modulation of Blimp-1 occurring during IFN-α-driven DC differentiation. Of note, monocyte-derived IFN-α DC and in vitro IFN-α-treated pDC shared a restricted pattern of miRNAs regulating Blimp-1 expression as well as some similar phenotypic, molecular and functional hallmarks, supporting the existence of a potential relationship between these DC populations. On the whole, these data uncover a new role of Blimp-1 in human DC differentiation driven by IFN-α and identify Blimp-1 as an IFN-α-mediated key regulator potentially accounting for shared functional features between IFN-α DC and pDC.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/metabolismo , Interferon-alfa/farmacologia , MicroRNAs/genética , Monócitos/citologia , Proteínas Repressoras/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Dendríticas/efeitos dos fármacos , Perfilação da Expressão Gênica , Células HeLa , Humanos , Fenótipo , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa