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.
Front Cell Infect Microbiol ; 13: 1245299, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37953797

RESUMO

Staphylococcus aureus (S. aureus) is a pathogen associated with a wide variety of diseases, from minor to life-threatening infections. Antibiotic-resistant strains have emerged, leading to increasing concern about the control of S. aureus infections. The development of vaccines may be one way to overcome these resistant strains. However, S. aureus ability to internalize into cells - and thus to form a reservoir escaping humoral immunity - is a challenge for vaccine development. A role of T cells in the elimination of persistent S. aureus has been established in mice but it remains to be established if CD8+ T cells could display a cytotoxic activity against S. aureus infected cells. We examined in vitro the ability of CD8+ T cells to recognize and kill dendritic cells infected with S. aureus. We first evidenced that both primary mouse dendritic cells and DC2.4 cell line can be infected with S. aureus. We then generated a strain of S. aureus expressing a model CD8 epitope and transgenic F5 CD8+ T cells recognizing this model epitope were used as reporter T cells. In response to S. aureus-infected dendritic cells, F5 CD8+ T cells produced IFN-γ in an antigen-specific manner and displayed an increased ability to kill infected cells. Altogether, these results demonstrate that cells infected by S. aureus display bacteria-derived epitopes at their surface that are recognized by CD8+ T cells. This paves the way for the development of CD8+ T cell-based therapies against S. aureus.


Assuntos
Linfócitos T CD8-Positivos , Staphylococcus aureus , Camundongos , Animais , Linfócitos T Citotóxicos , Epitopos de Linfócito T , Células Dendríticas
2.
PLoS Pathog ; 14(8): e1007158, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30089163

RESUMO

Human papillomavirus type 16 (HPV16) and other oncoviruses have been shown to block innate immune responses and to persist in the host. However, to avoid viral persistence, the immune response attempts to clear the infection. IL-1ß is a powerful cytokine produced when viral motifs are sensed by innate receptors that are members of the inflammasome family. Whether oncoviruses such as HPV16 can activate the inflammasome pathway remains unknown. Here, we show that infection of human keratinocytes with HPV16 induced the secretion of IL-1ß. Yet, upon expression of the viral early genes, IL-1ß transcription was blocked. We went on to show that expression of the viral oncoprotein E6 in human keratinocytes inhibited IRF6 transcription which we revealed regulated IL-1ß promoter activity. Preventing E6 expression using siRNA, or using E6 mutants that prevented degradation of p53, showed that p53 regulated IRF6 transcription. HPV16 abrogation of p53 binding to the IRF6 promoter was shown by ChIP in tissues from patients with cervical cancer. Thus E6 inhibition of IRF6 is an escape strategy used by HPV16 to block the production IL-1ß. Our findings reveal a struggle between oncoviral persistence and host immunity; which is centered on IL-1ß regulation.


Assuntos
Regulação da Expressão Gênica/imunologia , Evasão da Resposta Imune/imunologia , Fatores Reguladores de Interferon/metabolismo , Interleucina-1beta/biossíntese , Infecções por Papillomavirus/imunologia , Papillomavirus Humano 16/imunologia , Humanos , Fatores Reguladores de Interferon/imunologia , Interleucina-1beta/imunologia , Queratinócitos/imunologia , Queratinócitos/metabolismo , Queratinócitos/virologia , Proteínas Oncogênicas Virais/metabolismo , Infecções por Papillomavirus/metabolismo , Proteínas Repressoras/metabolismo
3.
J Infect Dis ; 211(4): 571-81, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25006047

RESUMO

BACKGROUND: Bone and joint infection, mainly caused by Staphylococcus aureus, is associated with significant morbidity and mortality, characterized by severe inflammation and progressive bone destruction. Studies mostly focused on the interaction between S. aureus and osteoblasts, the bone matrix-forming cells, while interactions between S. aureus and osteoclasts, the only cells known to be able to degrade bone, have been poorly explored. METHODS: We developed an in vitro infection model of primary murine osteoclasts to study the direct impact of live S. aureus on osteoclastogenesis and osteoclast resorption activity. RESULTS: Staphylococcal infection of bone marrow-derived osteoclast precursors induced their differentiation into activated macrophages that actively secreted proinflammatory cytokines. These cytokines enhanced the bone resorption capacity of uninfected mature osteoclasts and promoted osteoclastogenesis of the uninfected precursors at the site of infection. Moreover, infection of mature osteoclasts by live S. aureus directly enhanced their ability to resorb bone by promoting cellular fusion. CONCLUSIONS: Our results highlighted two complementary mechanisms involved in bone loss during bone and joint infection, suggesting that osteoclasts could be a pivotal target for limiting bone destruction.


Assuntos
Reabsorção Óssea/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Osteoclastos/microbiologia , Osteoclastos/fisiologia , Staphylococcus aureus/patogenicidade , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Citocinas/metabolismo , Durapatita , Camundongos , Modelos Biológicos , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética
4.
J Immunol ; 193(7): 3398-408, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25194054

RESUMO

The stimulation of TLRs by pathogen-derived molecules leads to the production of proinflammatory cytokines. Because uncontrolled inflammation can be life threatening, TLR regulation is important; however, few studies have identified the signaling pathways that contribute to the modulation of TLR expression. In this study, we examined the relationship between activation and the transcriptional regulation of TLR9. We demonstrate that infection of primary human epithelial cells, B cells, and plasmacytoid dendritic cells with dsDNA viruses induces a regulatory temporary negative-feedback loop that blocks TLR9 transcription and function. TLR9 transcriptional downregulation was dependent on TLR9 signaling and was not induced by TLR5 or other NF-κB activators, such as TNF-α. Engagement of the TLR9 receptor induced the recruitment of a suppressive complex, consisting of NF-κBp65 and HDAC3, to an NF-κB cis element on the TLR9 promoter. Knockdown of HDAC3 blocked the transient suppression in which TLR9 function was restored. These results provide a framework for understanding the complex pathways involved in transcriptional regulation of TLR9, immune induction, and inflammation against viruses.


Assuntos
Infecções por Vírus de DNA/imunologia , Vírus de DNA/imunologia , Regiões Promotoras Genéticas/imunologia , Receptor Toll-Like 9/imunologia , Transcrição Gênica/imunologia , Animais , Infecções por Vírus de DNA/genética , Infecções por Vírus de DNA/patologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Histona Desacetilases/genética , Histona Desacetilases/imunologia , Humanos , Masculino , Camundongos , Células NIH 3T3 , Plasmócitos/imunologia , Plasmócitos/patologia , Receptor Toll-Like 9/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/imunologia , Transcrição Gênica/genética
5.
J Exp Med ; 210(7): 1369-87, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23752229

RESUMO

Human papillomavirus type 16 (HPV16) and other oncogenic viruses have been reported to deregulate immunity by suppressing the function of the double-stranded DNA innate sensor TLR9. However, the mechanisms leading to these events remain to be elucidated. We show that infection of human epithelial cells with HPV16 promotes the formation of an inhibitory transcriptional complex containing NF-κBp50-p65 and ERα induced by the E7 oncoprotein. The E7-mediated transcriptional complex also recruited the histone demethylase JARID1B and histone deacetylase HDAC1. The entire complex bound to a specific region on the TLR9 promoter, which resulted in decreased methylation and acetylation of histones upstream of the TLR9 transcriptional start site. The involvement of NF-κB and ERα in the TLR9 down-regulation by HPV16 E7 was fully confirmed in cervical tissues from human patients. Importantly, we present evidence that the HPV16-induced TLR9 down-regulation affects the interferon response which negatively regulates viral infection. Our studies highlight a novel HPV16-mediated mechanism that combines epigenetic and transcriptional events to suppress a key innate immune sensor.


Assuntos
Papillomavirus Humano 16/imunologia , Papillomavirus Humano 16/patogenicidade , Proteínas E7 de Papillomavirus/imunologia , Receptor Toll-Like 9/genética , Sequência de Bases , Linhagem Celular Tumoral , Colo do Útero/imunologia , Colo do Útero/metabolismo , Colo do Útero/virologia , Regulação para Baixo/genética , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Dados de Sequência Molecular , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/virologia , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo
6.
Immunity ; 35(2): 194-207, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21820332

RESUMO

Although Toll-like receptor 9 (TLR9) has been implicated in cytokine and type I interferon (IFN) production during malaria in humans and mice, the high AT content of the Plasmodium falciparum genome prompted us to examine the possibility that malarial DNA triggered TLR9-independent pathways. Over 6000 ATTTTTAC ("AT-rich") motifs are present in the genome of P. falciparum, which we show here potently induce type I IFNs. Parasite DNA, parasitized erythrocytes and oligonucleotides containing the AT-rich motif induce type I IFNs via a pathway that did not involve the previously described sensors TLR9, DAI, RNA polymerase-III or IFI16/p204. Rather, AT-rich DNA sensing involved an unknown receptor that coupled to the STING, TBK1 and IRF3-IRF7 signaling pathway. Mice lacking IRF3, IRF7, the kinase TBK1 or the type I IFN receptor were resistant to otherwise lethal cerebral malaria. Collectively, these observations implicate AT-rich DNA sensing via STING, TBK1 and IRF3-IRF7 in P. falciparum malaria.


Assuntos
Sequência Rica em At/genética , DNA de Protozoário/genética , Malária Falciparum/imunologia , Oligonucleotídeos/genética , Plasmodium falciparum/fisiologia , Animais , DNA de Protozoário/metabolismo , Perfilação da Expressão Gênica , Humanos , Imunidade Inata/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Malária Falciparum/parasitologia , Malária Falciparum/fisiopatologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Oligonucleotídeos/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas Serina-Treonina Quinases/metabolismo , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/genética
7.
Virology ; 417(2): 443-8, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21791360

RESUMO

HPV16 E6 deregulates G1/S cell cycle progression through p53 degradation preventing transcription of the CDK inhibitor p21(WAF1). However, additional mechanisms independent of p53 inactivation appear to exist. Here, we report that HPV16 E6 targets the cellular factor p150(Sal2), which positively regulates p21(WAF1) transcription. HPV16 E6 associates with p150(Sal2), inducing its functional inhibition by preventing its binding to cis elements on the p21(WAF1) promoter. A HPV16 E6 mutant, L110Q, which was unable to bind p150(Sal2), did not affect the ability of the cellular protein to bind p21(WAF1) promoter, underlining the linkage between these events. These data describe a novel mechanism by which HPV16 E6 induces cell cycle deregulation with a p53-independent pathway. The viral oncoprotein targets p150(Sal2), a positive transcription regulator of p21(WAF1) gene, preventing G1/S arrest and allowing cellular proliferation and efficient viral DNA replication.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/antagonistas & inibidores , Interações Hospedeiro-Patógeno , Proteínas Oncogênicas Virais/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Transcrição Gênica , Substituição de Aminoácidos/genética , Ciclo Celular , Linhagem Celular , Proteínas de Ligação a DNA , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Proteínas Oncogênicas Virais/genética , Proteínas Repressoras/genética
8.
J Immunol ; 185(11): 6439-47, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20980631

RESUMO

EBV infects most of the human population and is associated with a number of human diseases including cancers. Moreover, evasion of the immune system and chronic infection is an essential step for EBV-associated diseases. In this paper, we show that EBV can alter the regulation and expression of TLRs, the key effector molecules of the innate immune response. EBV infection of human primary B cells resulted in the inhibition of TLR9 functionality. Stimulation of TLR9 on primary B cells led to the production of IL-6, TNF-α, and IgG, which was inhibited in cells infected with EBV. The virus exerts its inhibitory function by decreasing TLR9 mRNA and protein levels. This event was observed at early time points after EBV infection of primary cells, as well as in an immortalized lymphoblastoid cell line. We determined that the EBV oncoprotein latent membrane protein 1 (LMP1) is a strong inhibitor of TLR9 transcription. Overexpression of LMP1 in B cells reduced TLR9 promoter activity, mRNA, and protein levels. LMP1 mutants altered in activating the NF-κB pathway prevented TLR9 promoter deregulation. Blocking the NF-κB pathway recovered TLR9 promoter activity. Mutating the NF-κB cis element on the TLR9 promoter restored luciferase transcription in the presence of LMP1. Finally, deletion of the LMP1 gene in the EBV genome abolished the ability of the virus to induce TLR9 downregulation. Our study describes a mechanism used by EBV to suppress the host immune response by deregulating the TLR9 transcript through LMP1-mediated NF-κB activation.


Assuntos
Regulação para Baixo/imunologia , Herpesvirus Humano 4/imunologia , Receptor Toll-Like 9/antagonistas & inibidores , Receptor Toll-Like 9/genética , Proteínas da Matriz Viral/fisiologia , Linfócitos B/imunologia , Linfócitos B/virologia , Linhagem Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Infecções por Vírus Epstein-Barr/imunologia , Humanos , Evasão da Resposta Imune , Imunidade Inata , Proteínas Oncogênicas Virais/fisiologia , Receptor Toll-Like 9/biossíntese , Transcrição Gênica/imunologia
9.
J Biol Chem ; 285(45): 34773-80, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20829351

RESUMO

Toll-like receptor 8 (TLR8), which is expressed primarily in myeloid cells, plays a central role in initiating immune responses to viral single-stranded RNA. Despite the great interest in the field of TLR8 research, very little is known in terms of TLR8 biology and its transcriptional regulation. Here, we describe the isolation of the hTLR8 promoter and the characterization of the molecular mechanisms involved in its regulation. Reporter gene analysis and ChIP assays demonstrated that the hTLR8 regulation of the basal transcription is regulated via three C/EBP cis-acting elements that required C/EBPδ and C/EBPß activity. In addition, we observed that R848 stimulation increases TLR8 transcriptional activity via an enhanced binding of C/EBPδ, and not C/EBPß, to its responsive sites within the TLR8 promoter. Moreover, we showed that IFN-γ also increased TLR8 transcription activity via the binding of STAT1 transcription factor to IFN-γ activated sequence elements on the TLR8 promoter and enhanced TLR8 functionality. These results shed new light on the mechanisms involved during TLR8-mediated innate immune response.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Elementos de Resposta/fisiologia , Fator de Transcrição STAT1/metabolismo , Receptor 8 Toll-Like/biossíntese , Transcrição Gênica/fisiologia , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/genética , Proteína delta de Ligação ao Facilitador CCAAT/imunologia , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Viral/imunologia , RNA Viral/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Receptor 8 Toll-Like/genética , Receptor 8 Toll-Like/imunologia
10.
J Immunol ; 184(3): 1168-79, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20042593

RESUMO

Human metapneumoviruses (HMPVs) are recently identified Paramyxoviridae that contribute to respiratory tract infections in children. No effective treatments or vaccines are available. Successful defense against virus infection relies on early detection by germ line-encoded pattern recognition receptors and activation of cytokine and type I IFN genes. Recently, the RNA helicase retinoic acid-inducible gene I (RIG-I) has been shown to sense HMPV. In this study, we investigated the abilities of two prototype strains of HMPV (A1 [NL\1\00] and B1 [NL\1\99]) to activate RIG-I and induce type I IFNs. Despite the abilities of both HMPV-A1 and HMPV-B1 to infect and replicate in cell lines and primary cells, only the HMPV-A1 strain triggered RIG-I to induce IFNA/B gene transcription. The failure of the HMPV-B1 strain to elicit type I IFN production was dependent on the B1 phosphoprotein, which specifically prevented RIG-I-mediated sensing of HMPV viral 5' triphosphate RNA. In contrast to most cell types, plasmacytoid dendritic cells displayed a unique ability to sense both HMPV-A1 and HMPV-B1 and in this case sensing was via TLR7 rather than RIG-I. Collectively, these data reveal differential mechanisms of sensing for two closely related viruses, which operate in cell type-specific manners.


Assuntos
RNA Helicases DEAD-box/metabolismo , Metapneumovirus/imunologia , Fosfoproteínas/metabolismo , Receptor 7 Toll-Like/metabolismo , Interferência Viral/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteína DEAD-box 58 , RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/fisiologia , Regulação Viral da Expressão Gênica/imunologia , Humanos , Imunidade Inata , Interferon-alfa/biossíntese , Interferon-alfa/genética , Interferon beta/biossíntese , Interferon beta/genética , Ligantes , Metapneumovirus/genética , Metapneumovirus/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/metabolismo , Infecções por Paramyxoviridae/virologia , Fosfoproteínas/genética , RNA Viral/genética , Receptores Imunológicos , Especificidade da Espécie , Receptor 7 Toll-Like/deficiência , Receptor 7 Toll-Like/fisiologia , Células Vero
11.
Proc Natl Acad Sci U S A ; 106(14): 5789-94, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19297619

RESUMO

Malaria-induced sepsis is associated with an intense proinflammatory cytokinemia for which the underlying mechanisms are poorly understood. It has been demonstrated that experimental infection of humans with Plasmodium falciparum primes Toll-like receptor (TLR)-mediated proinflammatory responses. Nevertheless, the relevance of this phenomenon during natural infection and, more importantly, the mechanisms by which malaria mediates TLR hyperresponsiveness are unclear. Here we show that TLR responses are boosted in febrile patients during natural infection with P. falciparum. Microarray analyses demonstrated that an extraordinary percentage of the up-regulated genes, including genes involving TLR signaling, had sites for IFN-inducible transcription factors. To further define the mechanism involved in malaria-mediated "priming," we infected mice with Plasmodium chabaudi. The human data were remarkably predictive of what we observed in the rodent malaria model. Malaria-induced priming of TLR responses correlated with increased expression of TLR mRNA in a TLR9-, MyD88-, and IFNgamma-dependent manner. Acutely infected WT mice were highly susceptible to LPS-induced lethality while TLR9(-/-), IL12(-/-) and to a greater extent, IFNgamma(-/-) mice were protected. Our data provide unprecedented evidence that TLR9 and MyD88 are essential to initiate IL12 and IFNgamma responses and favor host hyperresponsiveness to TLR agonists resulting in overproduction of proinflammatory cytokines and the sepsis-like symptoms of acute malaria.


Assuntos
Imunidade Inata , Interferon gama/imunologia , Interleucina-12/imunologia , Malária/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Receptores Toll-Like/imunologia , Animais , Citocinas , Febre , Perfilação da Expressão Gênica , Humanos , Inflamação , Camundongos , Plasmodium chabaudi , Plasmodium falciparum , Sepse/parasitologia , Sepse/patologia , Receptor Toll-Like 9/imunologia , Receptores Toll-Like/genética , Fatores de Transcrição , Regulação para Cima/genética
12.
J Immunol ; 181(2): 1333-44, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18606688

RESUMO

TLR9 is critical in parasite recognition and host resistance to experimental infection with Trypanosoma cruzi. However, no information is available regarding nucleotide sequences and cellular events involved on T. cruzi recognition by TLR9. In silico wide analysis associated with in vitro screening of synthetic oligonucleotides demonstrates that the retrotransposon VIPER elements and mucin-like glycoprotein (TcMUC) genes in the T. cruzi genome are highly enriched for CpG motifs that are immunostimulatory for mouse and human TLR9, respectively. Importantly, infection with T. cruzi triggers high levels of luciferase activity under NF-kappaB-dependent transcription in HEK cells cotransfected with human TLR9, but not in control (cotransfected with human MD2/TLR4) HEK cells. Further, we observed translocation of TLR9 to the lysosomes during invasion/uptake of T. cruzi parasites by dendritic cells. Consistently, potent proinflammatory activity was observed when highly unmethylated T. cruzi genomic DNA was delivered to the endo-lysosomal compartment of host cells expressing TLR9. Thus, together our results indicate that the unmethylated CpG motifs found in the T. cruzi genome are likely to be main parasite targets and probably become available to TLR9 when parasites are destroyed in the lysosome-fused vacuoles during parasite invasion/uptake by phagocytes.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Lisossomos/imunologia , NF-kappa B/metabolismo , Receptor Toll-Like 9/metabolismo , Trypanosoma cruzi/imunologia , Animais , Linhagem Celular , Ilhas de CpG/imunologia , Células Dendríticas/citologia , Interações Hospedeiro-Parasita , Humanos , Lisossomos/parasitologia , Camundongos , Camundongos Knockout , NF-kappa B/imunologia , Oligodesoxirribonucleotídeos/imunologia , Retroelementos , Receptor Toll-Like 9/imunologia , Trypanosoma cruzi/genética
13.
J Biol Chem ; 283(6): 3376-3384, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18057002

RESUMO

Innate immunity is the first line defense against invading pathogens. During Gram-negative bacterial infection, the Toll-like receptor 4 and MD-2 complex recognize lipopolysaccharide present in the bacterial cell wall. This recognition can be enhanced 100-1000-fold by CD14. However, the beneficial role provided by CD14 becomes detrimental in the context of sepsis and septic shock. An understanding of how CD14 functions will therefore benefit treatments targeted at both immune suppression and immune enhancement. In the present study, we use site-directed mutagenesis to address the role of disulfide bonds and N-linked glycosylation on CD14. A differential impact is observed for the five disulfide bonds on CD14 folding, with the first two (Cys(6)-Cys(17) and Cys(15)-Cys(32)) being indispensable, the third and fourth (Cys(168)-Cys(198) and Cys(222)-Cys(253)) being important, and the last (Cys(287)-Cys(333)) being dispensable. A functional role is observed for the first disulfide bond because the C6A substitution severely reduces the ability of CD14 to confer lipopolysaccharide responsiveness to U373 cells. Two of the four predicted glycosylation sites, asparagines 132 and 263, are actually involved in N-linked glycosylation, resulting in heterogeneity in CD14 molecular weight. Furthermore, glycosylation at Asn(132) plays a role in CD14 trafficking and upstream and/or downstream ligand interactions. When mapped onto the crystal structure of mouse CD14, the first two disulfide bonds and Asn(132) are in close proximity to the initial beta strands of the leucine rich repeat domain. Thus, disulfide bonds and N-linked glycosylation in the initial beta sheets of the inner concave surface of CD14 are crucial for structure and function.


Assuntos
Receptores de Lipopolissacarídeos/química , Receptores de Lipopolissacarídeos/fisiologia , Animais , Asparagina/química , Linhagem Celular Tumoral , Cristalografia por Raios X , Dissulfetos , Glicosilação , Humanos , Camundongos , Modelos Moleculares , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Temperatura
14.
Nat Immunol ; 8(5): 487-96, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17417641

RESUMO

Increased concentrations of DNA-containing immune complexes in the serum are associated with systemic autoimmune diseases such as lupus. Stimulation of Toll-like receptor 9 (TLR9) by DNA is important in the activation of plasmacytoid dendritic cells and B cells. Here we show that HMGB1, a nuclear DNA-binding protein released from necrotic cells, was an essential component of DNA-containing immune complexes that stimulated cytokine production through a TLR9-MyD88 pathway involving the multivalent receptor RAGE. Moreover, binding of HMGB1 to class A CpG oligodeoxynucleotides considerably augmented cytokine production by means of TLR9 and RAGE. Our data demonstrate a mechanism by which HMGB1 and RAGE activate plasmacytoid dendritic cells and B cells in response to DNA and contribute to autoimmune pathogenesis.


Assuntos
Proteína HMGB1/fisiologia , Lúpus Eritematoso Sistêmico/imunologia , Oligodesoxirribonucleotídeos/imunologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Receptor Toll-Like 9/metabolismo , Animais , Complexo Antígeno-Anticorpo , Linfócitos B , Ilhas de CpG , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Proteína HMGB1/biossíntese , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor para Produtos Finais de Glicação Avançada/biossíntese , Receptores de Superfície Celular/metabolismo , Receptor Toll-Like 9/fisiologia
15.
Proc Natl Acad Sci U S A ; 104(6): 1919-24, 2007 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-17261807

RESUMO

Hemozoin (HZ) is an insoluble crystal formed in the food vacuole of malaria parasites. HZ has been reported to induce inflammation by directly engaging Toll-like receptor (TLR) 9, an endosomal receptor. "Synthetic" HZ (beta-hematin), typically generated from partially purified extracts of bovine hemin, is structurally identical to natural HZ. When HPLC-purified hemin was used to synthesize the crystal, beta-hematin had no inflammatory activity. In contrast, natural HZ from Plasmodium falciparum cultures was a potent TLR9 inducer. Natural HZ bound recombinant TLR9 ectodomain, but not TLR2. Both TLR9 stimulation and TLR9 binding of HZ were abolished by nuclease treatment. PCR analysis demonstrated that natural HZ is coated with malarial but not human DNA. Purified malarial DNA activated TLR9 but only when DNA was targeted directly to the endosome with a transfection reagent. Stimulatory quantities of natural HZ contain <1 microg of malarial DNA; its potency in activating immune responses was even greater than transfecting malarial DNA. Thus, although the malarial genome is extremely AT-rich, its DNA is highly proinflammatory, with the potential to induce cytokinemia and fever during disease. However, its activity depends on being bound to HZ, which we propose amplifies the biological responses to malaria DNA by targeting it to a TLR9(+) intracellular compartment.


Assuntos
Apresentação de Antígeno , DNA de Protozoário/metabolismo , Hemeproteínas/fisiologia , Imunidade Inata , Plasmodium falciparum/genética , Receptor Toll-Like 9/metabolismo , Animais , DNA de Protozoário/imunologia , Humanos , Ativação Linfocitária/imunologia , Melanoma Experimental , Camundongos , Plasmodium falciparum/imunologia , Receptor Toll-Like 9/imunologia
16.
Microbes Infect ; 8(9-10): 2432-41, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16859951

RESUMO

Broad immune responses, in particular specific for the NS3 protein and mediated by both CD8+ and CD4+T lymphocytes, are thought to play a critical role in the control of hepatitis C virus (HCV) infection. In this study, we searched for novel HLA-B*0702 NS3 restricted epitopes following an optimized NS3NS4 immunization protocol in transgenic mice expressing HLA-B*0702 molecule. Combining predicted and overlapping peptides, we identified two novel epitopes, WPA10 (aa 1111-1120) and LSP10 (aa 1153-1162), which triggered significant IFN-gamma-producing T cell frequencies and high CTL responses. Both epitopes were shown to be immunogenic when used as synthetic peptides to immunize mice. The relevance of these epitopes to humans was demonstrated, as both were able in vitro to recall specific IFN-gamma and IL10-producing cells from peripheral blood mononuclear cells of HCV infected patients. Such epitopes enlarge the pool of NS3-specific CD8+T cell epitopes available to perform immunomonitoring of HCV infection and to develop vaccines.


Assuntos
Antígenos HLA-B/imunologia , Hepacivirus/imunologia , Hepatite C/imunologia , Hepatite C/virologia , Proteínas não Estruturais Virais/imunologia , Alelos , Sequência de Aminoácidos , Animais , Linfócitos T CD8-Positivos/imunologia , Sequência Conservada , Mapeamento de Epitopos , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Antígenos HLA-B/genética , Antígeno HLA-B7 , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinação , Vacinas contra Hepatite Viral/imunologia , Proteínas não Estruturais Virais/genética
17.
J Virol ; 78(19): 10460-9, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15367612

RESUMO

In vitro studies have described the synthesis of an alternative reading frame form of the hepatitis C virus (HCV) core protein that was named F protein or ARFP (alternative reading frame protein) and includes a domain coded by the +1 open reading frame of the RNA core coding region. The expression of this protein in HCV-infected patients remains controversial. We have analyzed peripheral blood from 47 chronically or previously HCV-infected patients for the presence of T lymphocytes and antibodies specific to the ARFP. Anti-ARFP antibodies were detected in 41.6% of the patients infected with various HCV genotypes. Using a specific ARFP 99-amino-acid polypeptide as well as four ARFP predicted class I-restricted 9-mer peptides, we show that 20% of the patients display specific lymphocytes capable of producing gamma interferon, interleukin-10, or both cytokines. Patients harboring three different viral genotypes (1a, 1b, and 3) carried T lymphocytes reactive to genotype 1b-derived peptides. In longitudinal analysis of patients receiving therapy, both core and ARFP-specific T-cell- and B-cell-mediated responses were documented. The magnitude and kinetics of the HCV antigen-specific responses differed and were not linked with viremia or therapy outcome. These observations provide strong and new arguments in favor of the synthesis, during natural HCV infection, of an ARFP derived from the core sequence. Moreover, the present data provide the first demonstration of the presence of T-cell-mediated immune responses directed to this novel HCV antigen.


Assuntos
Hepacivirus/imunologia , Hepatite C/imunologia , Memória Imunológica , Linfócitos T/imunologia , Proteínas do Core Viral/imunologia , Adulto , Idoso , Sequência de Aminoácidos , Antígenos Virais/biossíntese , Antígenos Virais/imunologia , Antivirais/uso terapêutico , Linfócitos B/imunologia , Hepacivirus/efeitos dos fármacos , Hepacivirus/genética , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Anticorpos Anti-Hepatite C/sangue , Humanos , Interferon-alfa/uso terapêutico , Interferon gama/biossíntese , Interleucina-10/biossíntese , Pessoa de Meia-Idade , Dados de Sequência Molecular , Ribavirina/uso terapêutico , Homologia de Sequência , Resultado do Tratamento , Proteínas do Core Viral/biossíntese , Proteínas do Core Viral/genética , Viremia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...