Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Eur J Immunol ; 48(2): 250-257, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28980305

RESUMO

Viruses, particularly the Epstein-Barr virus (EBV) has long been suspected to exacerbate acute arthritic symptoms. However, the cell populations that contribute to import viruses into the inflamed tissues remain to be identified. In the present study, we have investigated the role of monocytes in the transport of Murid herpesvirus 68 (MHV-68), a mouse virus closely related to EBV, using the serum transfer-induced arthritis (STIA) model. We found compelling evidence that MHV-68 infection markedly increased disease severity in NR4A1-/- mice, which are deficient for Ly6Clow monocytes. In contrast, the MHV-68-induced enhancement of joint inflammation was lessened in CCR2-/- mice, suggesting the involvement of inflammatory Ly6Chigh monocytes in viral transport. We also observed that following selective depletion of monocyte subsets by administration of clodronate, MHV-68 transport into the synovium occurs only in the presence of Ly6Chigh monocytes. Tracking of adoptively transferred Ly6Chigh GFP infected monocytes into arthritic CCR2-/- mice by two-photon intravital microscopy showed that this monocyte subset has the capacity to deliver viruses to inflamed AR joints, as confirmed by the detection of viral DNA in inflamed tissues of recipient mice. We thus conclude that Ly6Chigh monocytes import MHV-68 when they are mobilized to the inflamed arthritic joint.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 4/fisiologia , Monócitos/imunologia , Rhadinovirus/fisiologia , Infecções Tumorais por Vírus/imunologia , Transferência Adotiva , Animais , Antígenos Ly/metabolismo , Artrite Experimental/virologia , Artrite Reumatoide/virologia , Células Cultivadas , DNA Viral/análise , Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/virologia , Feminino , Infecções por Herpesviridae/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/transplante , Muridae , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Receptores CCR2/genética , Rhadinovirus/patogenicidade , Infecções Tumorais por Vírus/virologia
2.
PLoS One ; 11(12): e0168034, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27930721

RESUMO

CCR2 is a chemokine receptor expressed on the surface of blood leukocytes, particularly «Ly6Chi¼ inflammatory monocytes and microglia. Signaling through this receptor is thought to influence the immune activity of microglia as well as monocytes egress from the bone marrow (BM) and their trafficking into the central nervous system (CNS) in several neurological diseases. During experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE), CCR2 deficiency has been reported to exacerbate the outcome of the disease. However, the precise contribution of CCR2 expressed in cells of the CNS or peripheral monocytes in the protection against HSE remains unclear. To dissect the differential role of CCR2 during HSE, chimeric mice with receptor deficiency in the brain or blood cells were generated by transplanting wild-type (WT) C57BL/6 or CCR2-/- BM-derived cells in CCR2-/- (WT→CCR2-/-) and WT (CCR2-/-→WT) mice, respectively. Our results indicate that following intranasal infection with 1.2x106 plaque forming units of HSV-1, CCR2 deficiency in hematopoietic cells and, to a lesser extent, in CNS exacerbates the outcome of HSE. Mortality rates of CCR2-/- (71.4%) and CCR2-/-→WT (57.1%) mice were significantly higher than that of WT (15.3%; P<0.01 and P<0.05, respectively) but the difference did not reach statistical significance for WT→CCR2-/- animals (42.8%; P = 0.16). Both peripheral and CNS deficiencies in CCR2 resulted in increased infectious viral titers and wider dissemination of HSV antigens in the brain as well as an overproduction of inflammatory cytokines and chemokines including IL-1ß, IL-6, CCL2, CCL3 and CCL5. Furthermore, CCR2 deficiency in the hematopoietic system altered monocytes egress from the BM and their recruitment to the CNS, which may contribute to the failure in HSV-1 containment. Collectively, these data suggest that CCR2 expressed on cells of CNS and especially on peripheral monocytes is important for the control of HSV-1 replication and inflammatory environment during experimental HSE.


Assuntos
Encéfalo/imunologia , Encefalite por Herpes Simples/imunologia , Células-Tronco Hematopoéticas/imunologia , Herpesvirus Humano 1/imunologia , Receptores CCR2/deficiência , Animais , Química Encefálica/imunologia , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/imunologia , Microglia/metabolismo , Monócitos/imunologia , Receptores CCR2/fisiologia , Carga Viral/imunologia
3.
PLoS One ; 10(10): e0139856, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26444420

RESUMO

Leukotriene B4 (LTB4), a central mediator of inflammation, is well known for its chemoattractant properties on effectors cells of the immune system. LTB4 also has the ability to control microbial infection by improving host innate defenses through the release of antimicrobial peptides and modulation of intracellular Toll-like receptors (TLRs) expression in response to agonist challenge. In this report, we provide evidences that LTB4 acts on nucleotide-binging oligomerization domain 2 (NOD2) pathway to enhance immune response against influenza A infection. Infected mice receiving LTB4 show improved survival, lung architecture and reduced lung viral loads as compared to placebo-treated animals. NOD2 and its downstream adaptor protein IPS-1 have been found to be essential for LTB4-mediated effects against IAV infection, as absence of NOD2 or IPS-1 diminished its capacity to control viral infection. Treatment of IAV-infected mice with LTB4 induces an increased activation of IPS-1-IRF3 axis leading to an enhanced production of IFNß in lungs of infected mice. LTB4 also has the ability to act on the RICK-NF-κB axis since administration of LTB4 to mice challenged with MDP markedly increases the secretion of IL-6 and TNFα in lungs of mice. TAK1 appears to be essential to the action of LTB4 on NOD2 pathway since pretreatment of MEFs with TAK1 inhibitor prior stimulation with IAV or MDP strongly abrogated the potentiating effects of LTB4 on both IFNß and cytokine secretion. Together, our results demonstrate that LTB4, through its ability to activate TAK1, potentiates both IPS-1 and RICK axis of the NOD2 pathway to improve host innate responses.


Assuntos
Imunidade Inata/imunologia , Leucotrieno B4/imunologia , Proteína Adaptadora de Sinalização NOD2/imunologia , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Inflamação/imunologia , Fator Regulador 3 de Interferon/imunologia , Interferon beta/imunologia , Interleucina-6/imunologia , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/microbiologia , MAP Quinase Quinase Quinases/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , Transdução de Sinais/imunologia
4.
Int Immunol ; 24(11): 693-704, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22843747

RESUMO

Pattern-recognition receptors such as Toll-like receptors (TLRs) are essential sensors implicated in the early and efficient innate immune response against pathogens. We have previously demonstrated that leukotriene B(4)(LTB(4)) has the capacity to enhance leukocyte responses to TLR9 ligands and to control viral infection. In this report, we provide evidence that LTB(4) treatment of human neutrophils leads to a potentiation in proinflammatory cytokine secretion induced by various myeloid differentiation factor 88-dependent TLR agonists. LTB(4) failed to enhance TLR mRNA levels as well as expression of TLR2 and TLR4 receptors, suggesting that LTB(4) acts through intracellular mechanism(s) to potentiate neutrophil responses to TLR ligands. We found that while IRAK can be activated by LTB(4), this process is dispensable to LTB(4) to potentiate neutrophil responses to TLR ligands since pretreatment of neutrophils with IRAK1/4 inhibitor did not affect its potentiating effects. However, our data clearly show that LTB(4) treatment of neutrophils led to the phosphorylation of downstream signaling molecules, TAK1 and p38, a process found essential to observe an increased secretion of cytokines by neutrophils activated with TLR ligands. Pretreatment of neutrophils with TAK1 or p38 kinase inhibitors strongly repressed the effect of LTB(4) on cytokine synthesis by neutrophils stimulated with LTA, LPS or CpG. The same pattern was observed in agonist-treated human embryonic kidney 293 cells transfected with TAK1-targeting siRNA where secretion of IL-8 was significantly reduced to basal levels. These results indicate that TAK1 and p38 kinases appear to be central in the 'priming effect' of LTB(4) on neutrophils to enhance response to TLR ligands.


Assuntos
Leucotrieno B4/imunologia , MAP Quinase Quinase Quinases/imunologia , Neutrófilos/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Western Blotting , Células Cultivadas , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Citometria de Fluxo , Células HEK293 , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/imunologia , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-8/imunologia , Interleucina-8/metabolismo , Leucotrieno B4/farmacologia , Ligantes , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Oligodesoxirribonucleotídeos/imunologia , Oligodesoxirribonucleotídeos/farmacologia , Inibidores de Proteínas Quinases/imunologia , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácidos Teicoicos/imunologia , Ácidos Teicoicos/farmacologia , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/agonistas , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Nature ; 481(7380): 199-203, 2011 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-22158124

RESUMO

The largest mucosal surface in the body is in the gastrointestinal tract, a location that is heavily colonized by microbes that are normally harmless. A key mechanism required for maintaining a homeostatic balance between this microbial burden and the lymphocytes that densely populate the gastrointestinal tract is the production and transepithelial transport of poly-reactive IgA (ref. 1). Within the mucosal tissues, B cells respond to cytokines, sometimes in the absence of T-cell help, undergo class switch recombination of their immunoglobulin receptor to IgA, and differentiate to become plasma cells. However, IgA-secreting plasma cells probably have additional attributes that are needed for coping with the tremendous bacterial load in the gastrointestinal tract. Here we report that mouse IgA(+) plasma cells also produce the antimicrobial mediators tumour-necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS), and express many molecules that are commonly associated with monocyte/granulocytic cell types. The development of iNOS-producing IgA(+) plasma cells can be recapitulated in vitro in the presence of gut stroma, and the acquisition of this multifunctional phenotype in vivo and in vitro relies on microbial co-stimulation. Deletion of TNF-α and iNOS in B-lineage cells resulted in a reduction in IgA production, altered diversification of the gut microbiota and poor clearance of a gut-tropic pathogen. These findings reveal a novel adaptation to maintaining homeostasis in the gut, and extend the repertoire of protective responses exhibited by some B-lineage cells.


Assuntos
Imunoglobulina A/imunologia , Intestino Delgado/citologia , Intestino Delgado/imunologia , Plasmócitos/citologia , Plasmócitos/imunologia , Animais , Células da Medula Óssea/citologia , Linhagem da Célula , Células Cultivadas , Quimera/imunologia , Citrobacter rodentium/imunologia , Técnicas de Cocultura , Feminino , Vida Livre de Germes , Granulócitos/citologia , Granulócitos/metabolismo , Imunidade Inata/imunologia , Imunoglobulina A/biossíntese , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Intestino Delgado/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Monócitos/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/metabolismo , Fenótipo , Plasmócitos/metabolismo , Baço/citologia , Células Estromais/citologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
6.
PLoS One ; 5(10): e13742, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-21060793

RESUMO

BACKGROUND: Toll-like receptors (TLRs) play a crucial role in the activation of innate immunity in response to many viruses. We previously reported the implication of TLR2 in the recognition of Epstein-Barr virus (EBV) by human monocytes. Because murine gammaherpesvirus-68 (MHV-68) is a useful model to study human gammaherpesvirus pathogenesis in vivo, we evaluated the importance of mouse TLR2 in the recognition of MHV-68. METHODOLOGY/PRINCIPAL FINDINGS: In studies using transfected HEK293 cells, MHV-68 lead to the activation of NF-κB reporter through TLR2. In addition, production of interleukin-6 (IL-6) and interferon-α (IFN-α) upon MHV-68 stimulation was reduced in murine embryonic fibroblasts (MEFs) derived from TLR2-/- and MyD88-/- mice as compared to their wild type (WT) counterpart. In transgenic mice expressing a luciferase reporter gene under the control of the mTLR2 promoter, MHV-68 challenge activated TLR2 transcription. Increased expression levels of TLR2 on blood granulocytes (CD115(-)Gr1(+)) and inflammatory monocytes (CD115(+)Gr1(+)), which mobilized to the lungs upon infection with MHV-68, was also confirmed by flow cytometry. Finally, TLR2 or MyD88 deficiency was associated with decreased IL-6 and type 1 IFN production as well as increased viral burden during short-term challenges with MHV-68. CONCLUSIONS/SIGNIFICANCE: TLR2 contributes to the production of inflammatory cytokines and type 1 IFN as well as to the control of viral burden during infection with MHV-68. Taken together, our results suggest that the TLR2 pathway has a relevant role in the recognition of this virus and in the subsequent activation of the innate immune response.


Assuntos
Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/fisiopatologia , Receptor 2 Toll-Like/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Gammaherpesvirinae/isolamento & purificação , Genes Reporter , Infecções por Herpesviridae/virologia , Humanos , Interferon gama/biossíntese , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Receptor 2 Toll-Like/genética
7.
PLoS One ; 5(7): e11908, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20689596

RESUMO

BACKGROUND: Epstein-Barr virus is recognized to cause lymphoproliferative disorders and is also associated with cancer. Evidence suggests that monocytes are likely to be involved in EBV pathogenesis, especially due to a number of cellular functions altered in EBV-infected monocytes, a process that may affect efficient host defense. Because type I interferons (IFNs) are crucial mediators of host defense against viruses, we investigated the effect of EBV infection on the IFNalpha pathway in primary human monocytes. METHODOLOGY/PRINCIPAL FINDINGS: Infection of monocytes with EBV induced IFNalpha secretion but inhibited the positive feedback loop for the amplification of IFNalpha. We showed that EBV infection induced the expression of suppressor of cytokine signaling 3 (SOCS3) and, to a lesser extent, SOCS1, two proteins known to interfere with the amplification of IFNalpha secretion mediated by the JAK/STAT signal transduction pathway. EBV infection correlated with a blockage in the activation of JAK/STAT pathway members and affected the level of phosphorylated IFN regulatory factor 7 (IRF7). Depletion of SOCS3, but not SOCS1, by small interfering RNA (siRNA) abrogated the inhibitory effect of EBV on JAK/STAT pathway activation and significantly restored IFNalpha secretion. Finally, transfection of monocytes with the viral protein Zta caused the upregulation of SOCS3, an event that could not be recapitulated with mutated Zta. CONCLUSIONS/SIGNIFICANCE: We propose that EBV protein Zta activates SOCS3 protein as an immune escape mechanism that both suppresses optimal IFNalpha secretion by human monocytes and favors a state of type I IFN irresponsiveness in these cells. This immunomodulatory effect is important to better understand the aspects of the immune response to EBV.


Assuntos
Herpesvirus Humano 4/patogenicidade , Interferon-alfa/metabolismo , Monócitos/metabolismo , Monócitos/virologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Western Blotting , Linhagem Celular , Células Cultivadas , Citometria de Fluxo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
8.
J Immunol ; 185(6): 3620-31, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20713890

RESUMO

TLR9 plays an important role in innate defense against viruses by the detection of CpG motifs of foreign DNA within intracellular compartments. In this study, we evaluated the ability of EBV to promote monocyte and plasmacytoid dendritic cell (pDC) activation and cytokine release through TLR9 activation. We demonstrated that treatment of primary monocytes with EBV and with purified EBV DNA induced the release of IL-8 through TLR9. Activation of TLR9 by viral DNA requires endosomal maturation because pretreatment of monocytes with chloroquine strongly reduced IL-8 secretion. However, pretreatment of monocytes with siRNA directed against TLR2, with inhibitory ODN (iODN) or with a combination of both inhibitors strongly reduced the secretion of IL-8, providing evidence of a dual action of TLR2 and TLR9 in EBV recognition by monocytes. In contrast, production of MCP-1 and IL-10 in EBV-treated monocytes was mainly regulated through TLR2. Although EBV does not establish infection in pDCs, challenge with either live EBV particles or isolated EBV DNA was found to induce the release of IFN-alpha through TLR9, as supported by blockage of TLR9 activity with iODN or chloroquine. The role of TLR9 in the recognition of EBV by pDCs appears to be dominant, as confirmed by the marked inhibitory effect of iODN observed on the synthesis of IFN-alpha, IL-6, and IL-8 by pDCs. These results demonstrate that recognition of EBV by TLR9 is differently orchestrated in primary monocytes and pDCs to optimize viral recognition and antiviral response.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Herpesvirus Humano 4/imunologia , Monócitos/imunologia , Monócitos/virologia , Receptor Toll-Like 9/fisiologia , Comunicação Celular/genética , Comunicação Celular/imunologia , Células Cultivadas , Citocinas/biossíntese , Células Dendríticas/metabolismo , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/patologia , Regulação da Expressão Gênica/imunologia , Herpesvirus Humano 4/patogenicidade , Humanos , Mediadores da Inflamação/fisiologia , Monócitos/metabolismo , Transporte Proteico/genética , Transporte Proteico/imunologia , Receptor Toll-Like 9/biossíntese , Receptor Toll-Like 9/genética , Regulação para Cima/genética , Regulação para Cima/imunologia
9.
Ann Pathol ; 28(4): 321-3, 2008 Sep.
Artigo em Francês | MEDLINE | ID: mdl-18928875

RESUMO

We report a case of sclerosing angiomatoid nodular transformation of the spleen incidently discovered in a 41-year-old man. The macroscopic examination showed the presence of a reddish brown, well delineated but not encapsulated, multinodular lesion being histologically characterized by nodules made up of complex vascular structures lined by monomorphous but non atypical endothelial cells, surrounded by fibrin and a collagen stroma rich in spumous macrophages and hemosiderophages. The immunohistochemical markers carried out showed the presence of capillaries, veins and sinusoids normally found within the splenic parenchyma, but adopting an unusual configuration. This distinct entity, recently described and completely benign, must be included in the differential diagnosis of the vascular lesions of the spleen, which includes, among others, the hemangioma, the littoral cell angioma, the hemangioendothelioma and the inflammatory myofibroblastic tumor.


Assuntos
Hemangioendotelioma/patologia , Baço/patologia , Esplenopatias/patologia , Neoplasias Esplênicas/patologia , Idoso , Alcoolismo/complicações , Alcoolismo/patologia , Antígenos CD/análise , Capilares/patologia , Colágeno/análise , Hemangioendotelioma/cirurgia , Humanos , Imuno-Histoquímica , Masculino , Esclerose/patologia , Baço/irrigação sanguínea , Esplenectomia , Esplenopatias/cirurgia , Neoplasias Esplênicas/cirurgia , Veias/patologia , Vênulas/patologia
10.
J Virol ; 81(15): 8016-24, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17522215

RESUMO

Epstein-Barr virus (EBV) is a gammaherpesvirus infecting the majority of the human adult population in the world. TLR2, a member of the Toll-like receptor (TLR) family, has been implicated in the immune responses to different viruses including members of the herpesvirus family, such as human cytomegalovirus, herpes simplex virus type 1, and varicella-zoster virus. In this report, we demonstrate that infectious and UV-inactivated EBV virions lead to the activation of NF-kappaB through TLR2 using HEK293 cells cotransfected with TLR2-expressing vector along with NF-kappaB-Luc reporter plasmid. NF-kappaB activation in HEK293-TLR2 cells (HEK293 cells transfected with TLR2) by EBV was not enhanced by the presence of CD14. The effect of EBV was abrogated by pretreating HEK293-TLR2 cells with blocking anti-TLR2 antibodies or by preincubating viral particles with neutralizing anti-EBV antibodies 72A1. In addition, EBV infection of primary human monocytes induced the release of MCP-1 (monocyte chemotactic protein 1), and the use of small interfering RNA targeting TLR2 significantly reduced such a chemokine response to EBV. Taken together, these results indicate that TLR2 may be an important pattern recognition receptor in the immune response directed against EBV infection.


Assuntos
Quimiocina CCL2/metabolismo , Herpesvirus Humano 4/imunologia , Monócitos/imunologia , Monócitos/virologia , Receptor 2 Toll-Like/imunologia , Adulto , Linhagem Celular , Quimiocina CCL2/imunologia , Genes Reporter , Humanos , Receptores de Lipopolissacarídeos/imunologia , Monócitos/citologia , NF-kappa B/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor 2 Toll-Like/genética , Vírion/imunologia
11.
Virus Res ; 119(2): 134-45, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16545476

RESUMO

Epstein-Barr virus (EBV) is an oncogenic human herpesvirus that persistently infects approximately 90% of the world's population. Such a remarkably sustained of viral infectivity relies on EBV's ability to evade the host immune defenses. A crucial part of this anti-EBV response is mediated by cytotoxic CD8+ T lymphocytes, which maintain a life-long control over proliferating latently-infected B cells in order to prevent these from giving rise to lymphomatous diseases. On the other hand, little has been done to assess the role of phagocytes-mediated innate immunity in the pathogenesis of EBV infection. In the course of primary EBV infection, episodes of neutropenia and monocytopenia can be observed during the acute phase of infection. According to the role of those cells in the non specific and specific immunity, such a decrease in circulating phagocytes may then temporarily affect the immune defense and potentially influence the outcome of EBV infection. Recent studies have demonstrated that EBV infects both neutrophils and monocytes and modulates several of their biological functions. This review covers the current state of our knowledge relative to the role of neutrophils and monocytes in EBV pathogenesis and describes the nature of countermeasures deployed by EBV against these cells.


Assuntos
Infecções por Vírus Epstein-Barr/imunologia , Herpesvirus Humano 4/imunologia , Imunidade Inata , Fagócitos/imunologia , Fagócitos/virologia , Herpesvirus Humano 4/patogenicidade , Humanos , Monócitos/imunologia , Monócitos/virologia , Neutrófilos/imunologia , Neutrófilos/virologia
12.
J Immunol ; 174(3): 1587-93, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15661920

RESUMO

Human CMV is often associated with transplant rejection and opportunistic infections such as pneumonia in immunosuppressed patients. Current anti-CMV therapies, although effective, show relatively high toxicity, which seriously limits their long-term use. In this study, we provide evidence that leukotriene B(4) (LTB(4)) plays an important role in the fight against murine CMV (MCMV) infection in vivo. Intravenous administration of 50 and 500 ng/kg/day of LTB(4) to mice infected with a lethal dose of MCMV significantly increases their survival (50 and 70%, respectively), compared with the placebo-treated group (10% of survival). In mice infected with a sublethal dose of MCMV and treated daily with 50 ng/kg/day of LTB(4), the salivary gland viral loads were found to be reduced by 66% compared with the control group. Furthermore, using an allogeneic bone marrow transplantation mouse model, the frequency of MCMV reactivation from latently infected mice was much lower (38%) in LTB(4) (500 ng/kg)-treated mice than in the placebo-treated group (78%). Finally, in experiments using 5-lipoxygenase-deficient mice, MCMV viral loads in salivary glands were found to be higher in animals unable to produce leukotrienes than in the control groups, supporting a role of endogenous 5-lipoxygenase products, possibly LTB(4), in host defense against CMV infection.


Assuntos
Antivirais/uso terapêutico , Transplante de Medula Óssea/efeitos adversos , Infecções por Citomegalovirus/prevenção & controle , Leucotrieno B4/uso terapêutico , Muromegalovirus/fisiologia , Ativação Viral , Latência Viral , Animais , Antivirais/deficiência , Antivirais/genética , Antivirais/fisiologia , Araquidonato 5-Lipoxigenase/deficiência , Araquidonato 5-Lipoxigenase/genética , Araquidonato 5-Lipoxigenase/fisiologia , Transplante de Medula Óssea/imunologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/mortalidade , Infecções por Citomegalovirus/virologia , Feminino , Rejeição de Enxerto/genética , Rejeição de Enxerto/prevenção & controle , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/prevenção & controle , Injeções Intravenosas , Leucotrieno B4/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Glândulas Salivares/efeitos dos fármacos , Glândulas Salivares/virologia , Baço/citologia , Baço/transplante , Baço/virologia , Carga Viral , Ativação Viral/genética , Ativação Viral/imunologia , Latência Viral/genética , Latência Viral/imunologia
13.
J Infect Dis ; 189(11): 2001-9, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15143466

RESUMO

BACKGROUND: CD8(+) T cells can control human immunodeficiency virus (HIV) through the lysis of infected cells and the release of soluble mediators, such as macrophage inflammatory protein (MIP)-1 beta, which prevent entry of HIV and/or inhibit HIV replication. Because neutrophils represent a major source of alpha-defensins and, to a lesser extent, MIP-1 beta, we determined whether leukotriene B(4) (LTB(4)), a potent neutrophil agonist, would trigger the release of these 2 anti-HIV peptides. METHODS: Plasma samples from HIV-uninfected subjects receiving intravenous bolus of LTB(4) were analyzed for alpha-defensins and MIP-1 beta levels by use of enzyme-linked immunosorbent assay. Furthermore, in vitro analysis of intracellular and secreted levels of alpha-defensins of resting and LTB(4)-activated neutrophils from HIV-uninfected and HIV-infected subjects were determined. LTB(4) modulation of CD63 and CD66b markers associated with degranulation were studied by use of flow cytometry. Chemotaxis of neutrophils from HIV-uninfected and HIV-infected subjects toward LTB(4) or interleukin (IL)-8 was determined by use of migration assays. RESULTS: Administration of LTB(4) to humans caused a dose-dependent plasmatic increase in alpha-defensins and MIP-1 beta proteins, with peak levels observed 2 h after administration of LTB(4). Neutrophils isolated from HIV-infected and HIV-uninfected subjects contained similar levels of stored alpha-defensins that were effectively secreted in vitro, in response to LTB(4) activation. Chemotaxis of neutrophils toward LTB(4) or IL-8 was identical among the groups of subjects. CONCLUSION: LTB(4) induced the secretion alpha-defensins and MIP-1 beta. Neutrophils from HIV-infected subjects were fully responsive to LTB(4), which highlights a potential usefulness of this lipid mediator in the management of HIV infection.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , Leucotrieno B4/farmacologia , Proteínas Inflamatórias de Macrófagos/sangue , Neutrófilos/imunologia , alfa-Defensinas/sangue , Antígenos CD/imunologia , Antígenos de Neoplasias/imunologia , Moléculas de Adesão Celular/imunologia , Quimiocina CCL4 , Quimiotaxia/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Proteínas Ligadas por GPI , Infecções por HIV/sangue , Infecções por HIV/tratamento farmacológico , Humanos , Interleucina-8/imunologia , Interleucina-8/farmacologia , Leucotrieno B4/imunologia , Proteínas Inflamatórias de Macrófagos/imunologia , Proteínas Inflamatórias de Macrófagos/metabolismo , Neutrófilos/metabolismo , Glicoproteínas da Membrana de Plaquetas/imunologia , Tetraspanina 30 , alfa-Defensinas/imunologia , alfa-Defensinas/metabolismo
14.
J Gen Virol ; 85(Pt 5): 1319-1328, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15105549

RESUMO

The human herpesvirus 6 (HHV-6) immediate-early (IE) 1 protein undergoes SUMOylation events during the infectious process. In the present work, we report that Lys-802 (K-802) of IE1 from HHV-6 variant B is the only target residue capable of conjugation to SUMO-1/SMT3C/Sentrin-1, SUMO-2/SMT3A/Sentrin-3 or SUMO-3/SMT3B/Sentrin-2 as determined by transfection and in vitro SUMOylation experiments. PolySUMOylated forms of IE1 were also observed, suggesting that SUMO branching occurs at the K-802 residue. Overexpression of SUMO-1, -2 and -3 led to an overall increase in IE1 levels, irrespective of K-802. The SUMO residues could be efficiently removed by incubating SUMOylated IE1 with SENP1, a recently identified SUMO peptidase. SUMOylation-deficient mutants of IE1 co-localized with nuclear promyelocytic leukaemia protein (PML) oncogenic domains (PODs) as efficiently as WT IE1, indicating that POD targeting is independent of IE1 SUMOylation status. However, in contrast to infection, PODs did not aggregate in IE1B-transfected cells, suggesting that other viral proteins are involved in the process. Transactivation studies indicated that IE1, in combination with IE2, could efficiently transactivate diverse promoters, independent of its SUMOylation status. Overall, the results presented provide a detailed biochemical characterization of post-translational modifications of the HHV-6 IE1 protein by SUMO peptides, contributing to our understanding of the complex interactions between herpesviruses and the SUMO-conjugation pathway.


Assuntos
Herpesvirus Humano 6/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Linhagem Celular , Proteínas Imediatamente Precoces/genética , Lisina , Mutação , Fosfoproteínas/genética , Estrutura Terciária de Proteína
15.
J Biol Chem ; 277(27): 24148-54, 2002 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-11971896

RESUMO

Infection of human monocytes by Epstein-Barr virus (EBV) has been linked to a decrease in the production of proinflammatory mediators as well as an impairment of phagocytosis. Considering the key role of protein kinases C (PKCs) in many biological functions of monocytes, including phagocytosis, we investigated the effects of EBV on the PKC activity in infected monocytes. Our results indicate that infection of monocytes by EBV impairs both phorbol 12-myristate 13-acetate (PMA)-induced translocation of PKC isozymes alpha and beta from cytosol to membrane as well as the PKC enzymatic activity. Similarly, the subcellular distribution of the receptor for activated C kinase (RACK), an anchoring protein essential to PKC translocation, was also found to be reduced in EBV-infected monocytes. Transfection of 293T cells with an expression vector coding for the immediate-early protein ZEBRA of EBV resulted in impaired PMA-induced translocation and activity of PKC. Using co-immunoprecipitation assays, the ZEBRA protein was found to physically interact with the RACK1 protein. Thus interaction of ZEBRA with RACK likely results in the inhibition of PKC activity, which in turn affects functions of monocytes, such as phagocytosis.


Assuntos
Herpesvirus Humano 4/fisiologia , Monócitos/fisiologia , Fagocitose/fisiologia , Proteína Quinase C/sangue , Acetato de Tetradecanoilforbol/farmacologia , Proteínas Virais , Membrana Celular/enzimologia , Citosol/enzimologia , Proteínas de Ligação a DNA/sangue , Inibidores Enzimáticos/farmacologia , Humanos , Indóis/farmacologia , Monócitos/virologia , Naftalenos/farmacologia , Fagocitose/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Transporte Proteico , Transativadores/sangue
16.
J Biol Chem ; 277(22): 19679-87, 2002 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-11901159

RESUMO

Immediate-early (IE) proteins are the first proteins expressed following viral entry and play a crucial role in the initiation of infection. We report the cloning and characterization of a full-length IE1 transcript and protein (IE1B) from human herpesvirus 6 (HHV-6) variant B. The IE1B transcript consists of five exons (3720 nucleotides), three of which are coding for the IE1 protein. The 1078-amino acid-long IE1B protein is 62% identical and 75% similar to the 941-amino acid IE1 from HHV-6 variant A. IE1B protein can be detected at 4 h post-infection (P.I.), and it is distributed as small intranuclear structures. The maximal number of IE1 bodies ( approximately 10-12/nucleus) is detected at 12 h P.I. after which the IE1 bodies condense into 1-3 larger entities by 24-48 h P.I. During infection the IE1B protein is phosphorylated on serine and threonine residues. IE1B undergoes further post-translational modification with its conjugation to the small ubiquitin-like modifier (SUMO-1) peptide. IE1B colocalizes with SUMO-1 and promyelocytic leukemia nuclear bodies during infection as well as in transfection experiments. Finally, IE1 from variant B is a weaker transactivator than IE1 from variant A, when assayed using heterologous promoters. Overall, the characterization of the HHV-6 IE1B protein presented highlights the similarity and divergence between IE1 from both variants and provides useful information pertaining to the early phase of infection.


Assuntos
Núcleo Celular/metabolismo , Herpesvirus Humano 6/metabolismo , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Sistema Livre de Células , Clonagem Molecular , DNA Complementar/metabolismo , Éxons , Vetores Genéticos , Glutationa Transferase/metabolismo , Humanos , Cinética , Modelos Genéticos , Dados de Sequência Molecular , Fosforilação , Regiões Promotoras Genéticas , Proteína da Leucemia Promielocítica , Ligação Proteica , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , RNA/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Serina/química , Espectrometria de Fluorescência , Treonina/química , Fatores de Tempo , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA