Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 17(9): e1009878, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34473805

RESUMO

SARS-CoV-2 fine-tunes the interferon (IFN)-induced antiviral responses, which play a key role in preventing coronavirus disease 2019 (COVID-19) progression. Indeed, critically ill patients show an impaired type I IFN response accompanied by elevated inflammatory cytokine and chemokine levels, responsible for cell and tissue damage and associated multi-organ failure. Here, the early interaction between SARS-CoV-2 and immune cells was investigated by interrogating an in vitro human peripheral blood mononuclear cell (PBMC)-based experimental model. We found that, even in absence of a productive viral replication, the virus mediates a vigorous TLR7/8-dependent production of both type I and III IFNs and inflammatory cytokines and chemokines, known to contribute to the cytokine storm observed in COVID-19. Interestingly, we observed how virus-induced type I IFN secreted by PBMC enhances anti-viral response in infected lung epithelial cells, thus, inhibiting viral replication. This type I IFN was released by plasmacytoid dendritic cells (pDC) via an ACE-2-indipendent but Neuropilin-1-dependent mechanism. Viral sensing regulates pDC phenotype by inducing cell surface expression of PD-L1 marker, a feature of type I IFN producing cells. Coherently to what observed in vitro, asymptomatic SARS-CoV-2 infected subjects displayed a similar pDC phenotype associated to a very high serum type I IFN level and induction of anti-viral IFN-stimulated genes in PBMC. Conversely, hospitalized patients with severe COVID-19 display very low frequency of circulating pDC with an inflammatory phenotype and high levels of chemokines and pro-inflammatory cytokines in serum. This study further shed light on the early events resulting from the interaction between SARS-CoV-2 and immune cells occurring in vitro and confirmed ex vivo. These observations can improve our understanding on the contribution of pDC/type I IFN axis in the regulation of the anti-viral state in asymptomatic and severe COVID-19 patients.


Assuntos
COVID-19/imunologia , Células Dendríticas/classificação , Interferon Tipo I/metabolismo , SARS-CoV-2/imunologia , Adulto , Idoso de 80 Anos ou mais , Infecções Assintomáticas , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Células Dendríticas/virologia , Células Epiteliais/citologia , Feminino , Hospitalização , Humanos , Interferon Tipo I/imunologia , Pulmão/citologia , Masculino , Pessoa de Meia-Idade , Neuropilina-1/metabolismo , Fenótipo , Índice de Gravidade de Doença , Receptor 7 Toll-Like/metabolismo
2.
PLoS Pathog ; 17(4): e1009505, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33857267

RESUMO

The Tick-borne encephalitis virus (TBEV) causes different disease symptoms varying from asymptomatic infection to severe encephalitis and meningitis suggesting a crucial role of the human host immune system in determining the fate of the infection. There is a need to understand the mechanisms underpinning TBEV-host interactions leading to protective immunity. To this aim, we studied the response of human peripheral blood mononuclear cells (PBMC) to the whole formaldehyde inactivated TBEV (I-TBEV), the drug substance of Encepur, one of the five commercially available vaccine. Immunophenotyping, transcriptome and cytokine profiling of PBMC revealed that I-TBEV generates differentiation of a sub-population of plasmacytoid dendritic cells (pDC) that is specialized in type I interferon (IFN) production. In contrast, likely due to the presence of aluminum hydroxide, Encepur vaccine was a poor pDC stimulus. We demonstrated I-TBEV-induced type I IFN together with Interleukin 6 and BAFF to be critical for B cell differentiation to plasmablasts as measured by immunophenotyping and immunoglobulin production. Robust type I IFN secretion was induced by pDC with the concerted action of both viral E glycoprotein and RNA mirroring previous data on dual stimulation of pDC by both S. aureus and influenza virus protein and nucleic acid that leads to a type I IFN-mediated sustained immune response. E glycoprotein neutralization or high temperature denaturation and inhibition of Toll-like receptor 7 signalling confirmed the importance of preserving the functional integrity of these key viral molecules during the inactivation procedure and manufacturing process to produce a vaccine able to stimulate strong immune responses.


Assuntos
Células Dendríticas/imunologia , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/prevenção & controle , Interações entre Hospedeiro e Microrganismos , Interferon Tipo I/metabolismo , Vacinas Virais/imunologia , Antivirais/imunologia , Diferenciação Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/virologia , Encefalite Transmitida por Carrapatos/virologia , Humanos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , RNA Viral/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
3.
Leukemia ; 33(1): 132-147, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29946193

RESUMO

Cancer cells subvert host immune surveillance by altering immune checkpoint (IC) proteins. Some Epstein-Barr virus (EBV)-associated tumors have higher Programmed Cell Death Ligand, PD-L1 expression. However, it is not known how EBV alters ICs in the context of its preferred host, the B lymphocyte and in derived lymphomas. Here, we found that latency III-expressing Burkitt lymphoma (BL), diffuse large B-cell lymphomas (DLBCL) or their EBNA2-transfected derivatives express high PD-L1. In a DLBCL model, EBNA2 but not LMP1 is sufficient to induce PD-L1. Latency III-expressing DLBCL biopsies showed high levels of PD-L1. The PD-L1 targeting oncosuppressor microRNA miR-34a was downregulated in EBNA2-transfected lymphoma cells. We identified early B-cell factor 1 (EBF1) as a repressor of miR-34a transcription. Short hairpin RNA (shRNA)-mediated knockdown of EBF1 was sufficient to induce miR-34a transcription, which in turn reduced PD-L1. MiR-34a reconstitution in EBNA2-transfected DLBCL reduced PD-L1 expression and increased its immunogenicity in mixed lymphocyte reactions (MLR) and in three-dimensional biomimetic microfluidic chips. Given the importance of PD-L1 inhibition in immunotherapy and miR-34a dysregulation in cancers, our findings may have important implications for combinatorial immunotherapy, which include IC inhibiting antibodies and miR-34a, for EBV-associated cancers.


Assuntos
Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/metabolismo , Infecções por Vírus Epstein-Barr/complicações , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/imunologia , Linfoma Difuso de Grandes Células B/imunologia , MicroRNAs/genética , Proteínas Virais/metabolismo , Antígeno B7-H1/genética , Biomarcadores Tumorais/genética , Infecções por Vírus Epstein-Barr/virologia , Antígenos Nucleares do Vírus Epstein-Barr/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/virologia , Prognóstico , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Células Tumorais Cultivadas , Proteínas Virais/genética
4.
J Infect Dis ; 213(4): 674-83, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26386427

RESUMO

CD8(+) T cells specific to caspase-cleaved antigens derived from apoptotic T cells represent a principal player in chronic immune activation. Here, we found that both apoptotic epitope-specific and hepatitis C virus (HCV)-specific CD8(+) T cells were mostly confined within the effector memory (EM) or terminally differentiated EM CD45RA(+) cell subsets expressing a dysfunctional T-helper 1-like signature program in chronic HCV infection. However, apoptotic epitope-specific CD8(+) T cells produced tumor necrosis factor α and interleukin 2 at the intrahepatic level significantly more than HCV-specific CD8(+) T cells, despite both populations expressing high levels of programmed death 1 receptor. Contextually, only apoptotic epitope-specific CD8(+) T cells correlated with both interferon-stimulated gene levels in T cells and hepatic fibrosis score. Together, these data suggest that, compared with HCV-specific CD8(+) T cells, apoptotic epitope-specific CD8(+) T cells can better sustain chronic immune activation, owing to their capacity to produce tumor necrosis factor α, and exhibit greater resistance to inhibitory signals during chronic HCV infection.


Assuntos
Apoptose , Linfócitos T CD8-Positivos/imunologia , Hepatite C Crônica/imunologia , Hepatite C Crônica/patologia , Interferons/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Adulto , Idoso , Feminino , Humanos , Interleucina-2/metabolismo , Cirrose Hepática/patologia , Masculino , Pessoa de Meia-Idade , Fator de Necrose Tumoral alfa/metabolismo
5.
Sci Rep ; 5: 17078, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26602835

RESUMO

Modern strategies to develop vaccines against Mycobacterium tuberculosis (Mtb) aim to improve the current Bacillus Calmette-Guerin (BCG) vaccine or to attenuate the virulence of Mtb vaccine candidates. In the present study, the impact of wild type or mutated region of difference 1 (RD1) variants on the immunogenicity of Mtb and BCG recombinants was investigated in human primary dendritic cells (DC). A comparative analysis of transcriptome, signalling pathway activation, maturation, apoptosis, cytokine production and capacity to promote Th1 responses demonstrated that DC sense quantitative and qualitative differences in the expression of RD1-encoded factors--ESAT6 and CFP10--within BCG or Mtb backgrounds. Expansion of IFN-γ producing T cells was promoted by BCG::RD1-challenged DC, as compared to their BCG-infected counterparts. Although Mtb recombinants acted as a strong Th-1 promoting stimulus, even with RD1 deletion, the attenuated Mtb strain carrying a C-terminus truncated ESAT-6 elicited a robust Th1 promoting phenotype in DC. Collectively, these studies indicate a necessary but not sufficient role for the RD1 locus in promoting DC immune-regulatory functions. Additional mycobacterial factors are likely required to endow DC with a high Th1 polarizing capacity, a desirable attribute for a successful control of Mtb infection.


Assuntos
Proteínas de Bactérias/imunologia , Células Dendríticas/metabolismo , Mycobacterium tuberculosis/metabolismo , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Apoptose , Vacina BCG/genética , Vacina BCG/imunologia , Vacina BCG/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/imunologia , Perfilação da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutagênese , Fenótipo , Células Th1/citologia , Células Th1/imunologia , Células Th1/metabolismo , Fator de Transcrição RelA/metabolismo , Transcriptoma , Vacinas Sintéticas/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
PLoS Pathog ; 9(4): e1003220, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23592979

RESUMO

It has long been known that multiple sclerosis (MS) is associated with an increased Epstein-Barr virus (EBV) seroprevalence and high immune reactivity to EBV and that infectious mononucleosis increases MS risk. This evidence led to postulate that EBV infection plays a role in MS etiopathogenesis, although the mechanisms are debated. This study was designed to assess the prevalence and magnitude of CD8+ T-cell responses to EBV latent (EBNA-3A, LMP-2A) and lytic (BZLF-1, BMLF-1) antigens in relapsing-remitting MS patients (n = 113) and healthy donors (HD) (n = 43) and to investigate whether the EBV-specific CD8+ T cell response correlates with disease activity, as defined by clinical evaluation and gadolinium-enhanced magnetic resonance imaging. Using HLA class I pentamers, lytic antigen-specific CD8+ T cell responses were detected in fewer untreated inactive MS patients than in active MS patients and HD while the frequency of CD8+ T cells specific for EBV lytic and latent antigens was higher in active and inactive MS patients, respectively. In contrast, the CD8+ T cell response to cytomegalovirus did not differ between HD and MS patients, irrespective of the disease phase. Marked differences in the prevalence of EBV-specific CD8+ T cell responses were observed in patients treated with interferon-ß and natalizumab, two licensed drugs for relapsing-remitting MS. Longitudinal studies revealed expansion of CD8+ T cells specific for EBV lytic antigens during active disease in untreated MS patients but not in relapse-free, natalizumab-treated patients. Analysis of post-mortem MS brain samples showed expression of the EBV lytic protein BZLF-1 and interactions between cytotoxic CD8+ T cells and EBV lytically infected plasma cells in inflammatory white matter lesions and meninges. We therefore propose that inability to control EBV infection during inactive MS could set the stage for intracerebral viral reactivation and disease relapse.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Herpesvirus Humano 4/imunologia , Esclerose Múltipla Recidivante-Remitente/imunologia , Esclerose Múltipla Recidivante-Remitente/virologia , Transativadores/metabolismo , Adulto , Anticorpos Monoclonais Humanizados/uso terapêutico , Encéfalo/virologia , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/virologia , Feminino , Humanos , Interferon beta/uso terapêutico , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Natalizumab , Estudos Soroepidemiológicos , Adulto Jovem
7.
J Infect Dis ; 205(9): 1425-35, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22457295

RESUMO

BACKGROUND: Autophagy inhibits survival of intracellular Mycobacterium tuberculosis when induced by rapamycin or interferon γ (IFN-γ), but it remains unclear whether M. tuberculosis itself can induce autophagy and whether T cells play a role in M. tuberculosis-mediated autophagy. The aim of this study was to evaluate the impact of M. tuberculosis on autophagy in human primary macrophages and the role of specific T cells in this process. METHODS: M. tuberculosis (H37Rv)-infected macrophages were incubated with naive or M. tuberculosis-specific T cells. Autophagy was evaluated at 4 hours and 8 hours after infection by analyzing the levels of LC3-II (a hallmark of autophagy) and p62 (a protein degraded by autophagy). M. tuberculosis survival was evaluated by counting the colony-forming units. RESULTS: M. tuberculosis infection of macrophages inhibited the autophagic process at 8 hours after infection. Naive T cells could not rescue this block, whereas M. tuberculosis-specific T cells restored autophagy degradation, accompanied by enhanced bacterial killing. Notably, the effect of M. tuberculosis-specific T cells was not affected by neutralization of endogenous IFN-γ and tumor necrosis factor α and was blocked by preventing contact between macrophages and T cells, suggesting that cell-cell interaction is crucial. CONCLUSIONS: M. tuberculosis inhibits autophagy in human primary macrophages, and specific T cells can restore functional autophagic flux through cell-cell contact.


Assuntos
Autofagia/imunologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Linfócitos T/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Western Blotting , Comunicação Celular , Humanos , Interferon gama/antagonistas & inibidores , Interferon gama/metabolismo , Macrófagos/imunologia , Microscopia Confocal , Mycobacterium tuberculosis/patogenicidade , Fagossomos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteína Sequestossoma-1 , Células-Tronco/metabolismo , Linfócitos T/metabolismo , Tuberculose/metabolismo , Tuberculose/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
8.
Eur J Immunol ; 37(12): 3499-508, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17985330

RESUMO

Type I IFN are cytokines which play a central role in host resistance to viral or microbial infections and are important components linking innate and adaptive immunity. We and others have previously demonstrated that the production of IFN-beta by DC following bacterial infections or TLR triggering influences, in an autocrine manner, their maturation. In this study, we investigated whether IFN-beta release modulates the phenotype of the immature DC and their response to a subsequent TLR stimulation. The induction of CD86, HLA-DR, CD38 and B7H1 and the absence of CCR7 and CD83 expression upon IFN-beta treatment suggest that IFN-beta-primed DC remain at the site of infection acquiring an activated phenotype. These results prompted us to investigate the response of IFN-beta-primed DC to TLR stimulation. While IFN-beta pretreatment increases slightly the expression of maturation markers in TLR2- or TLR4-stimulated DC, it is able to modulate selectively the secretion of inflammatory and immuno-regulating cytokines. Interestingly, IL-27p28 subunit was induced by IFN-beta alone or during LPS-induced maturation of DC in a type I IFN-dependent manner through IFN regulatory factor-1 (IRF-1) activation. Taken together, our results shed light on the capacity of IFN-beta to finely tune DC response to invading pathogens.


Assuntos
Células Dendríticas/efeitos dos fármacos , Fator Regulador 1 de Interferon/fisiologia , Interferon beta/farmacologia , Interleucinas/biossíntese , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Antígenos CD/biossíntese , Antígenos CD/genética , Antígeno B7-2/biossíntese , Antígeno B7-2/genética , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunoglobulinas/biossíntese , Imunoglobulinas/genética , Interleucina-10/metabolismo , Interleucina-12/biossíntese , Interleucina-12/genética , Interleucina-6/metabolismo , Interleucinas/genética , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Subunidades Proteicas , Receptores CCR7/biossíntese , Receptores CCR7/genética , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Antígeno CD83
9.
J Biol Chem ; 281(36): 26188-95, 2006 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16849320

RESUMO

Here we identify Viperin as a highly inducible gene in response to lipopolysaccharide (LPS), double-stranded RNA (poly(I-C)) or Sendai virus (SV). The only known function of Viperin relates to its ability to inhibit human Cytomegalovirus replication. Very little data are available on the regulation of this gene. In silico analysis of the promoter identified two interferon (IFN)-stimulated response elements (ISRE), which in other genes bind IRF3 or the IFN-stimulated gene factor-3 (ISGF3) complex. LPS and poly(I-C) induce very high levels of Viperin in wild type cells but not in cells deficient in TRIF, TBK1, IRF3, or the type I IFNalpha/betaR. SV-induced Viperin gene expression was mediated independently of Toll-like receptor (TLR) signaling by retinoic acid-inducible gene (RIG-I) and the downstream adapter, mitochondrial anti-viral signaling (MAVS). Virus-induced Viperin expression was not attenuated in macrophages deficient in either TBK1 or IKKepsilon alone. Moreover, IRF3-deficient, but not IFNalpha/betaR deficient, macrophages still induced Viperin in response to SV. Promoter reporter studies combined with DNA immunoprecipitation assays identified the ISGF3 complex as the key regulator of Viperin gene expression. Moreover, positive regulatory domain I-binding factor 1 (PRDI-BF1, also called BLIMP1) binds the ISRE sites and competes with ISGF3 binding in a virus inducible manner to inhibit Viperin transcription. Collectively, these studies identify Viperin as a tightly regulated ISGF3 target gene, which is counter-regulated by PRDI-BF1.


Assuntos
Regulação da Expressão Gênica , Fator Gênico 3 Estimulado por Interferon/metabolismo , Proteínas/metabolismo , Proteínas Repressoras/metabolismo , Receptores Toll-Like/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Genes Reporter , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Gênico 3 Estimulado por Interferon/genética , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Fator 1 de Ligação ao Domínio I Regulador Positivo , Regiões Promotoras Genéticas , Proteínas/genética , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Proteínas Repressoras/genética , Receptores Toll-Like/genética , Fatores de Transcrição/genética , Transcrição Gênica
10.
Infect Immun ; 74(6): 3296-304, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16714557

RESUMO

The Mycobacterium tuberculosis genome encodes 13 sigma factors. We have previously shown that mutations in some of these transcriptional activators render M. tuberculosis sensitive to various environmental stresses and can attenuate the virulence phenotype. In this work, we focused on extracytoplasmic factor sigmaE and studied the effects induced by the deletion of its structural gene (sigE) in the infection of human monocyte-derived dendritic cells (MDDC). We found that the wild-type M. tuberculosis strain (H37Rv), the sigE mutant (ST28), and the complemented strain (ST29) were able to infect dendritic cells (DC) to similar extents, although at 4 days postinfection a reduced ability to grow inside MDDC was observed for the sigE mutant ST28. After mycobacterium capture, the majority of MDDC underwent full maturation and expressed both inflammatory cytokines, such as tumor necrosis factor alpha, and the regulatory cytokines interleukin-12 (IL-12), IL-18, and beta interferon (IFN-beta). Conversely, a higher level of production of IL-10 was observed in ST28-infected MDDC compared to H37Rv- or ST29-infected cell results. However, in spite of the presence of IL-10, supernatants from ST28-infected DC induced IFN-gamma production by T cells similarly to those from H37Rv-infected DC culture. On the other hand, IL-10 impaired CXCL10 production in sigE mutant-infected DC and, indeed, its neutralization restored CXCL10 secretion. In line with these results, supernatants from ST28-infected cells showed a decreased capability to recruit CXCR3+ CD4+ T cells compared to those obtained from H37Rv-infected DC culture. Thus, our findings suggest that the sigE mutant-induced secretion of IL-10 inhibits CXCL10 expression and, in turn, the recruitment of activated-effector cells involved in the formation of granulomas.


Assuntos
Proteínas de Bactérias/fisiologia , Quimiocinas CXC/biossíntese , Células Dendríticas/microbiologia , Interleucina-10/biossíntese , Mycobacterium tuberculosis/patogenicidade , Fator sigma/fisiologia , Linfócitos T/imunologia , Linfócitos T CD4-Positivos/fisiologia , Movimento Celular , Quimiocina CXCL10 , Quimiocinas/biossíntese , Humanos , Interferon gama/biossíntese , Interleucina-12/biossíntese , Interleucina-18/biossíntese , Fator de Necrose Tumoral alfa/biossíntese
11.
Infect Immun ; 74(3): 1480-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16495518

RESUMO

Aspergillus fumigatus is the most prevalent airborne fungal pathogen and causes fatal invasive aspergillosis in immunocompromised patients. Given the essential role of dendritic cells (DC) in initiating and regulating immune responses, we investigated the impact of A. fumigatus conidial infection on human DC. A. fumigatus conidia were rapidly internalized and induced the release of tumor necrosis factor alpha within the first 8 h. After A. fumigatus infection, the majority of DC underwent full maturation, although CCR7 expression was observed only in DC that had internalized the conidia. Additionally, the analysis of regulatory cytokines showed that infected DC simultaneously produced interleukin-12p70 (IL-12p70) and significant amounts of IL-10. IL-10 neutralization was not able to further increase IL-12p70 production from infected DC. Whereas the central role of IL-12 in the generation of Th1 cells has long been appreciated, recently two other members of the IL-12 family, IL-23 and IL-27, were reported to play important roles in the regulation of gamma interferon (IFN-gamma) production from naïve and memory T cells. A. fumigatus-infected DC were also able to express high levels of IL-23p19 and low levels of IL-27p28 at later stages of infection. According to this expression pattern, A. fumigatus-infected DC were able to prime IFN-gamma production of naïve T cells. Thus, this study on the expression of the new IL-12 family members controlling the Th1 response sheds light on a novel aspect of the contribution of DC to anti-Aspergillus immunity.


Assuntos
Aspergilose/imunologia , Aspergillus fumigatus/fisiologia , Citocinas/análise , Células Dendríticas/metabolismo , Receptores de Quimiocinas/metabolismo , Células Th1/imunologia , Aspergillus fumigatus/imunologia , Diferenciação Celular , Células Cultivadas/imunologia , Células Cultivadas/metabolismo , Células Dendríticas/microbiologia , Humanos , Interleucina-12/análise , Interleucina-12/metabolismo , Interleucina-17/análise , Interleucina-17/metabolismo , Interleucina-23 , Subunidade p19 da Interleucina-23 , Interleucinas/análise , Interleucinas/metabolismo , Ativação Linfocitária , Receptores CCR7 , Células Th1/efeitos dos fármacos
12.
Biochem J ; 377(Pt 2): 367-78, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14505489

RESUMO

IRFs [IFN (interferon) regulatory factors] constitute a family of transcription factors involved in IFN signalling and in the development and differentiation of the immune system. IRF-2 has generally been described as an antagonist of IRF-1-mediated transcription of IFN and IFN-inducible genes; however, it has been recently identified as a transcriptional activator of some genes, such as those encoding histone H4, VCAM-1 (vascular cell adhesion molecule-1) and Fas ligand. Biologically, IRF-2 plays an important role in cell growth regulation and has been shown to be a potential oncogene. Studies in knock-out mice have also implicated IRF-2 in the differentiation and functionality of haematopoietic cells. Here we show that IRF-2 expression in a myeloid progenitor cell line leads to reprogramming of these cells towards the megakaryocytic lineage and enables them to respond to thrombopoietin, as assessed by cell morphology and expression of specific differentiation markers. Up-regulation of transcription factors involved in the development of the megakaryocytic lineage, such as GATA-1, GATA-2, FOG-1 (friend of GATA-1) and NF-E2 (nuclear factor-erythroid-2), and transcriptional stimulation of the thrombopoietin receptor were also demonstrated. Our results provide evidence for a key role for IRF-2 in the induction of a programme of megakaryocytic differentiation, and reveal a remarkable functional diversity of this transcription factor in the regulation of cellular responses.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Megacariócitos/metabolismo , Proteínas Repressoras , Apoptose , Diferenciação Celular , Divisão Celular , Linhagem Celular , Linhagem da Célula , Humanos , Fator Regulador 2 de Interferon , Megacariócitos/citologia , Células Progenitoras Mieloides/fisiologia , Fatores de Transcrição/metabolismo , Ativação Transcricional
13.
J Leukoc Biol ; 74(3): 395-402, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12949243

RESUMO

Macrophages have a central role in innate-immune responses to bacteria. In the present work, we show that infection of human macrophages with Gram-positive pathogenic Streptococcus pyogenes or nonpathogenic Lactobacillus rhamnosus GG enhances mRNA expression of inflammatory chemokine ligands CCL2/monocyte chemoattractant protein-1 (MCP-1), CCL3/macrophage-inflammatory protein-1alpha (MIP-1alpha), CCL5/regulated on activation, normal T expressed and secreted, CCL7/MCP-3, CCL19/MIP-3beta, and CCL20/MIP-3alpha and CXC chemokine ligands CXCL8/interleukin (IL)-8, CXCL9/monokine induced by interferon-gamma (IFN-gamma), and CXCL10/IFN-inducible protein 10. Bacteria-induced CCL2, CCL7, CXCL9, and CXCL10 mRNA expression was partially dependent on ongoing protein synthesis. The expression of these chemokines and of CCL19 was dependent on bacteria-induced IFN-alpha/beta production. CCL19 and CCL20 mRNA expression was up-regulated by IL-1beta or tumor necrosis factor alpha (TNF-alpha), and in addition, IFN-alpha together with TNF-alpha further enhanced CCL19 gene expression. Synergy between IFN-alpha and TNF-alpha was also seen for CXCL9 and CXCL10 mRNA expression. Bacteria-stimulated macrophage supernatants induced the migration of T helper cell type 1 (Th1) cells, suggesting that in human macrophages, these bacteria can stimulate efficient inflammatory chemokine gene expression including those that recruit Th1 cells to the site of inflammation. Furthermore, L. rhamnosus-induced Th1 chemokine production could in part explain the proposed antiallergenic properties of this bacterium.


Assuntos
Quimiocinas/biossíntese , Quimiotaxia de Leucócito , Lactobacillus/fisiologia , Macrófagos/microbiologia , Streptococcus pyogenes/fisiologia , Células Th1/imunologia , Movimento Celular , Células Cultivadas , Quimiocinas/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Interferons/farmacologia , Macrófagos/metabolismo , RNA Mensageiro/análise
14.
J Immunol ; 170(3): 1174-82, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12538673

RESUMO

We recently reported that dendritic cells (DC) infected with Mycobacterium tuberculosis (Mtb) produce Th1/IFN-gamma-inducing cytokines, IFN-alpha beta and IL-12. In the present article, we show that maturing Mtb-infected DC express high levels of CCR7 and they become responsive to its ligand CCL21. Conversely, CCR5 expression was rapidly lost from the cell surface following Mtb infection. High levels of CCL3 and CCL4 were produced within 8 h after infection, which is likely to account for the observed CCR5 down-modulation on Mtb-infected DC. In addition, Mtb infection stimulated the secretion of CXCL9 and CXCL10. Interestingly, the synthesis of CXCL10 was mainly dependent on the Mtb-induced production of IFN-alpha beta. Indeed, IFN-alpha beta neutralization down-regulated CXCL10 expression, whereas the expression of CXCL9 appeared to be unaffected. The chemotactic activity of the Mtb-infected DC supernatants was evaluated by migration assays using activated NK, CD4(+), and CD8(+) cells that expressed both CCR5 and CXCR3. Mtb-induced expression of CCL3, CCL4, CXCL9, and CXCL10 was involved in the stimulation of NK and T cell migration. In accordance with the data on the IFN-alpha beta-induced expression of CXCL10, neutralization of IFN-alpha beta significantly reduced the chemotactic activity of the supernatant from Mtb-infected DC. This indicates that IFN-alpha beta may modulate the immune response through the expression of CXCL10, which along with CXCL9, CCL3, and CCL4 participates in the recruitment and selective homing of activated/effector cells, which are known to accumulate at the site of Mtb infection and take part in the formation of the granulomas.


Assuntos
Movimento Celular/genética , Quimiocinas CXC/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/imunologia , Interferon Tipo I/metabolismo , Células Matadoras Naturais/citologia , Ativação Linfocitária , Mycobacterium tuberculosis/imunologia , Subpopulações de Linfócitos T/citologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Células Cultivadas , Quimiocina CXCL10 , Quimiocinas/metabolismo , Quimiocinas/fisiologia , Quimiocinas CXC/genética , Técnicas de Cocultura , Células Dendríticas/microbiologia , Humanos , Interferon Tipo I/fisiologia , Ativação Linfocitária/genética , Receptores de Quimiocinas/biossíntese , Subpopulações de Linfócitos T/imunologia
15.
Oncogene ; 21(52): 7933-44, 2002 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-12439744

RESUMO

High expression of transferrin receptor (TfR) on the membrane of erythroid cells accounts for the high level of iron required to sustain heme synthesis. Several studies indicate that during erythroid differentiation TfR expression is highly dependent on transcriptional regulation. In this study we characterized the minimal region able to confer transcriptional regulation during erythroid differentiation in Friend leukemia cells (FLC). This region of 120 bp, upstream the transcription start site, contains an overlapping consensus recognition sequence for AP1/CREB/ATF transcription factors and for proteins of the Ets family and a GC rich region. Here, we report that both the Ets and the Ap1/CRE like sites are essential for promoter activity during erythroid differentiation. We showed that Ets-1 binds to the EBS-TfR and its binding activity decreases in FLC induced to differentiate and during normal erythroid differentiation. Consistent with this, FLC constitutively expressing Ets-1 show a decrease in TfR gene expression, globin mRNA and hemoglobin synthesis. We conclude that Ets-1 binding activity is modulated during erythroid maturation and that a deregulated expression of this transcription factor interferes with terminal erythroid differentiation.


Assuntos
Diferenciação Celular/fisiologia , Eritrócitos/química , Regulação da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores da Transferrina/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Sequência de Bases , Células Cultivadas , DNA , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Proto-Oncogênica c-ets-1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-ets , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
16.
J Biol Chem ; 277(25): 22377-85, 2002 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-11948194

RESUMO

Alveolar macrophages and newly recruited monocytes are targets of infection by Mycobacterium tuberculosis. Therefore, we examined the expression of interferon regulatory factor 1 (IRF-1), which plays an important role in host defense against M. tuberculosis, in undifferentiated and differentiated cells. Infection induced IRF-1 in both. IRF-1 from undifferentiated, uninfected monocytic cell lines was modified during extraction to produce specific species that were apparently smaller than intact IRF-1. After infection by M. tuberculosis or differentiation, intact IRF-1 was recovered. Subcellular fractions were assayed for the ability to modify IRF-1 or inhibit its modification. A serine protease on the cytoplasmic surface of an organelle or vesicle in the "lysosomal/mitochondrial" fraction from undifferentiated cells was responsible for the modification of IRF-1. Thus, the simplest explanation of the modification is cleavage of IRF-1 by the serine protease. Recovery of intact IRF-1 correlated with induction of a serine protease inhibitor that was able to significantly reduce the modification of IRF-1. The inhibitor was present in the cytoplasm of M. tuberculosis-infected or -differentiated cells. It is likely that induction of both IRF-1 and the serine protease inhibitor in response to infection by M. tuberculosis represent host defense mechanisms.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Monócitos/microbiologia , Mycobacterium tuberculosis/patogenicidade , Fosfoproteínas/biossíntese , Inibidores de Serina Proteinase/farmacologia , Apoptose , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Citoplasma/metabolismo , DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Immunoblotting , Fator Regulador 1 de Interferon , Cinética , Lisossomos/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Serina Endopeptidases/metabolismo , Frações Subcelulares
17.
Cytokine ; 17(1): 28-35, 2002 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-11886168

RESUMO

The anti-inflammatory cytokine interleukin 4 (IL-4) has shown both inductive and inhibitory effects on the replication of the human immunodeficiency virus type 1 (HIV-1) in primary CD4+ T cells and mononuclear phagocytes. In this study, IL-4 did not induce virus production, but inhibited phorbol esters (PMA)-stimulated HIV expression in chronically infected promonocytic U1 cells. This effect, however, was not accounted for by a decreased secretion of endogenous TNF-alpha induced by phorbol myristate acetate (PMA). We also observed that PMA upregulated the production of both IL-1beta and of IL-1 receptor antagonist (IL-1ra). IL-4 inhibited the secretion of IL-1beta and strongly increased that of IL-1ra; however, these effects were not responsible of IL-4-mediated inhibition of PMA-induced HIV expression since anti-IL-1ra antibodies did not revert IL-4 mediated suppression. U1 cells were transiently transfected with both wild-type (WT) long terminal repeat (LTR) constructs, or with LTR plasmids containing deletions of either the NF-kappaB or the Sp-1 binding sites. IL-4 inhibited LTR-driven transcription triggered by PMA stimulation of U1 cells, and this effect was dependent upon intact NF-kappaB but not Sp-1 binding sites. Thus, IL-4 may favour a state of microbiological quiescence in infected monocytic cells bypassing the induction of HIV expression mediated by pro-inflammatory cytokines.


Assuntos
HIV-1/metabolismo , Interleucina-1/metabolismo , Interleucina-4/farmacologia , Ésteres de Forbol/farmacologia , Sialoglicoproteínas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Sítios de Ligação , Linhagem Celular , Cloranfenicol O-Acetiltransferase/metabolismo , Regulação para Baixo , Humanos , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-4/metabolismo , Plasmídeos/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção
18.
Cell Signal ; 14(6): 537-45, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11897494

RESUMO

Macrophage cell lines exhibit different responses to IFN-gamma depending on their maturation stage. We investigated the mechanisms underlying the differential IFN-gamma responsiveness in the less mature P388.D1 and in mature RAW264.7 cells. A reduction in the binding activity of the signal transducer and activator of transcription-1 (STAT-1) to different STAT binding elements (SBEs) was observed in P388.D1. This reduced binding activity was not due to an impaired STAT-1 activation. Studies on the expression of a negative regulator of cytokine signalling, protein-inhibiting activated STAT-1 (PIAS-1), showed that this protein was expressed constitutively at high levels in P388.D1. Forced expression of a PIAS-1 homologue, the Gu binding protein (GBP), inhibited the STAT-1-mediated gene activation in RAW264.7 cells, whereas a construct expressing the 5' portion of GBP in the antisense orientation reverts the IFN-gamma-resistant phenotype of P388.D1. Thus, our results indicate that PIAS-1 may account for the differential IFN-gamma responsiveness in macrophage cell lines at different stages of maturation.


Assuntos
Interferon gama/antagonistas & inibidores , Macrófagos/imunologia , Proteínas/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , DNA Antissenso/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Interferon gama/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas Inibidoras de STAT Ativados , Proteínas/genética , RNA Mensageiro/biossíntese , Fator de Transcrição STAT1 , Transdução de Sinais , Transativadores/antagonistas & inibidores , Transativadores/metabolismo
19.
Blood Cells Mol Dis ; 29(3): 553-61, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12547252

RESUMO

Members of the Ets gene family are known to be expressed in the hematopoietic tissue and some of them play a pivotal role in normal hematopoietic cell development. Ets-1 gene expression was analyzed in Friend Leukemia Cells (FLC) induced to erythroid differentiation by DMSO. We show that the level of Ets-1 protein and its binding activity decreases in FLC along erythroid differentiation of primary human progenitors. The same behavior was observed during normal erythroid differentiation. Moreover, FLC constitutively expressing Ets-1 show a decrease in TfR gene expression, globin mRNA and hemoglobin synthesis. These data indicate that a decrease in Ets-1 binding activity is required for a normal erythroid maturation and that a deregulated expression of this transcription factor may interfere with terminal erythroid differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células Precursoras Eritroides/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Humanos , Células K562 , Proteína Proto-Oncogênica c-ets-1 , Proteínas Proto-Oncogênicas c-ets
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA