Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Viruses ; 15(6)2023 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-37376652

RESUMO

Macrophages are critical in the pathogenesis of a diverse group of viral pathogens, both as targets of infection and for eliciting primary defense mechanisms. Our prior in vitro work identified that CD40 signaling in murine peritoneal macrophages protects against several RNA viruses by eliciting IL-12, which stimulates the production of interferon gamma (IFN-γ). Here, we examine the role of CD40 signaling in vivo. We show that CD40 signaling is a critical, but currently poorly appreciated, component of the innate immune response using two distinct infectious agents: mouse-adapted influenza A virus (IAV, PR8) and recombinant VSV encoding the Ebola virus glycoprotein (rVSV-EBOV GP). We find that stimulation of CD40 signaling decreases early IAV titers, whereas loss of CD40 elevated early titers and compromised lung function by day 3 of infection. Protection conferred by CD40 signaling against IAV is dependent on IFN-γ production, consistent with our in vitro studies. Using rVSV-EBOV GP that serves as a low-biocontainment model of filovirus infection, we demonstrate that macrophages are a CD40-expressing population critical for protection within the peritoneum and T-cells are the key source of CD40L (CD154). These experiments reveal the in vivo mechanisms by which CD40 signaling in macrophages regulates the early host responses to RNA virus infection and highlight how CD40 agonists currently under investigation for clinical use may function as a novel class of broad antiviral treatments.


Assuntos
Antígenos CD40 , Infecções por Vírus de RNA , Vírus de RNA , Animais , Camundongos , Antígenos CD40/metabolismo , Interferon gama , Macrófagos , Infecções por Vírus de RNA/imunologia
2.
J Exp Med ; 220(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36515679

RESUMO

The longevity of plasma cells is dependent on their ability to access and reside in so-called niches that are predominantly located in the bone marrow. Here, by employing a traceable method to label recently generated plasma cells, we showed that homeostatic plasma cells in the bone marrow and spleen were continuously replenished by newly generated B220hiMHC-IIhi populations that progressively differentiated into B220loMHC-IIlo long-lived plasma cell (LLPC) populations. We also found that, in the bone marrow, germinal center (GC)-independent and GC-dependent plasma cells decayed similarly upon NP-CGG engagement, and both entered the B220loMHC-IIlo LLPC pool. Compared with NP+B220hiMHC-IIhi plasma cells, NP+B220loMHC-IIlo cells were more immobilized in the bone marrow niches and showed better survival potential. Thus, our results suggest that the adhesion status of bone marrow plasma cells is dynamically altered during their differentiation and is associated with provision of survival signals.


Assuntos
Medula Óssea , Plasmócitos , Plasmócitos/metabolismo , Diferenciação Celular , Células da Medula Óssea , Centro Germinativo , Sobrevivência Celular
3.
J Leukoc Biol ; 109(2): 309-325, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32441445

RESUMO

Many acute viral infections target tissue Mϕs, yet the mechanisms of Mϕ-mediated control of viruses are poorly understood. Here, we report that CD40 expressed by peritoneal Mϕs restricts early infection of a broad range of RNA viruses. Loss of CD40 expression enhanced virus replication as early as 12-24 h of infection and, conversely, stimulation of CD40 signaling with an agonistic Ab blocked infection. With peritoneal cell populations infected with the filovirus, wild-type (WT) Ebola virus (EBOV), or a BSL2 model virus, recombinant vesicular stomatitis virus encoding Ebola virus glycoprotein (rVSV/EBOV GP), we examined the mechanism conferring protection. Here, we demonstrate that restricted virus replication in Mϕs required CD154/CD40 interactions that stimulated IL-12 production through TRAF6-dependent signaling. In turn, IL-12 production resulted in IFN-γ production, which induced proinflammatory polarization of Mϕs, protecting the cells from infection. These CD40-dependent events protected mice against virus challenge. CD40-/- mice were exquisitely sensitive to intraperitoneal challenge with a dose of rVSV/EBOV GP that was sublethal to CD40+/+ mice, exhibiting viremia within 12 h of infection and rapidly succumbing to infection. This study identifies a previously unappreciated role for Mϕ-intrinsic CD40 signaling in controlling acute virus infection.


Assuntos
Antígenos CD40/metabolismo , Imunidade Inata , Macrófagos/imunologia , Macrófagos/virologia , Vírus de RNA/fisiologia , Transdução de Sinais , Viroses/imunologia , Replicação Viral/fisiologia , Doença Aguda , Animais , Ligante de CD40/metabolismo , Ebolavirus/fisiologia , Glicoproteínas/imunologia , Humanos , Interferon gama/metabolismo , Interleucina-12/biossíntese , Camundongos Endogâmicos C57BL , Modelos Biológicos , Peritônio/patologia , Peritônio/virologia , Fator 6 Associado a Receptor de TNF/metabolismo , Viroses/virologia
4.
PLoS Negl Trop Dis ; 13(6): e0006983, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31242184

RESUMO

BACKGROUND: T cell immunoglobulin mucin domain-1 (TIM-1) is a phosphatidylserine (PS) receptor, mediating filovirus entry into cells through interactions with PS on virions. TIM-1 expression has been implicated in Ebola virus (EBOV) pathogenesis; however, it remains unclear whether this is due to TIM-1 serving as a filovirus receptor in vivo or, as others have suggested, TIM-1 induces a cytokine storm elicited by T cell/virion interactions. Here, we use a BSL2 model virus that expresses EBOV glycoprotein to demonstrate the importance of TIM-1 as a virus receptor late during in vivo infection. METHODOLOGY/PRINCIPAL FINDINGS: Infectious, GFP-expressing recombinant vesicular stomatitis virus encoding either full length EBOV glycoprotein (EBOV GP/rVSV) or mucin domain deleted EBOV glycoprotein (EBOV GPΔO/rVSV) was used to assess the role of TIM-1 during in vivo infection. GFP-expressing rVSV encoding its native glycoprotein G (G/rVSV) served as a control. TIM-1-sufficient or TIM-1-deficient BALB/c interferon α/ß receptor-/- mice were challenged with these viruses. While G/rVSV caused profound morbidity and mortality in both mouse strains, TIM-1-deficient mice had significantly better survival than TIM-1-expressing mice following EBOV GP/rVSV or EBOV GPΔO/rVSV challenge. EBOV GP/rVSV or EBOV GPΔO/rVSV in spleen of infected animals was high and unaffected by expression of TIM-1. However, infectious virus in serum, liver, kidney and adrenal gland was reduced late in infection in the TIM-1-deficient mice, suggesting that virus entry via this receptor contributes to virus load. Consistent with higher virus loads, proinflammatory chemokines trended higher in organs from infected TIM-1-sufficient mice compared to the TIM-1-deficient mice, but proinflammatory cytokines were more modestly affected. To assess the role of T cells in EBOV GP/rVSV pathogenesis, T cells were depleted in TIM-1-sufficient and -deficient mice and the mice were challenged with virus. Depletion of T cells did not alter the pathogenic consequences of virus infection. CONCLUSIONS: Our studies provide evidence that at late times during EBOV GP/rVSV infection, TIM-1 increased virus load and associated mortality, consistent with an important role of this receptor in virus entry. This work suggests that inhibitors which block TIM-1/virus interaction may serve as effective antivirals, reducing virus load at late times during EBOV infection.


Assuntos
Ebolavirus/fisiologia , Doença pelo Vírus Ebola/virologia , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Ebolavirus/genética , Feminino , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Receptor Celular 1 do Vírus da Hepatite A/deficiência , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores Virais/deficiência , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Genética Reversa , Vesiculovirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
5.
Cell Rep ; 21(7): 1839-1852, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29141217

RESUMO

Effector T cells exhibiting features of either T helper 1 (Th1) or T follicular helper (Tfh) populations are essential to control experimental Plasmodium infection and are believed to be critical for resistance to clinical malaria. To determine whether Plasmodium-specific Th1- and Tfh-like effector cells generate memory populations that contribute to protection, we developed transgenic parasites that enable high-resolution study of anti-malarial memory CD4 T cells in experimental models. We found that populations of both Th1- and Tfh-like Plasmodium-specific memory CD4 T cells persist. Unexpectedly, Th1-like memory cells exhibit phenotypic and functional features of Tfh cells during recall and provide potent B cell help and protection following transfer, characteristics that are enhanced following ligation of the T cell co-stimulatory receptor OX40. Our findings delineate critical functional attributes of Plasmodium-specific memory CD4 T cells and identify a host-specific factor that can be targeted to improve resolution of acute malaria and provide durable, long-term protection against Plasmodium parasite re-exposure.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Imunidade Humoral , Memória Imunológica , Malária/imunologia , Plasmodium/imunologia , Células Th1/imunologia , Animais , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores OX40/metabolismo
6.
Methods Mol Biol ; 1325: 19-37, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26450376

RESUMO

Whole attenuated parasite vaccines designed to elicit immunity against the clinically silent preerythrocytic stage of Plasmodium infection represent the most efficacious experimental platforms currently in clinical trial. Studies in rodents and humans show that T cells mediate vaccine-induced protection. Thus, determining the quantitative and qualitative properties of these T cells remains a major research focus. Most rodent models of preerythrocytic anti-Plasmodium vaccination focus on circumsporozoite-specific CD8 T cell responses in BALB/c mice. However, CD4 T cells and non-circumsporozoite-specific CD8 T cells also significantly contribute to protection. Here we describe alternative approaches that enable detection and functional characterization of total CD8 and CD4 T cell responses induced by preerythrocytic vaccination in mice. These flow cytometry-based approaches rely on monitoring the modulation of expressed integrins and co-receptors on the surface of T cells in vaccinated mice. The approaches enable direct determination of the magnitude, kinetics, distribution, phenotype, and functional features of T cell responses induced by infection or whole-parasite vaccination using any mouse-parasite species combination.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/imunologia , Biologia Molecular/métodos , Vacinação/métodos , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Eritrócitos/imunologia , Humanos , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/uso terapêutico , Camundongos , Plasmodium/imunologia , Plasmodium/patogenicidade , Vacinas Atenuadas/imunologia
7.
Cell Host Microbe ; 17(5): 628-41, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25891357

RESUMO

The differentiation and protective capacity of Plasmodium-specific T cells are regulated by both positive and negative signals during malaria, but the molecular and cellular details remain poorly defined. Here we show that malaria patients and Plasmodium-infected rodents exhibit atypical expression of the co-stimulatory receptor OX40 on CD4 T cells and that therapeutic enhancement of OX40 signaling enhances helper CD4 T cell activity, humoral immunity, and parasite clearance in rodents. However, these beneficial effects of OX40 signaling are abrogated following coordinate blockade of PD-1 co-inhibitory pathways, which are also upregulated during malaria and associated with elevated parasitemia. Co-administration of biologics blocking PD-1 and promoting OX40 signaling induces excessive interferon-gamma that directly limits helper T cell-mediated support of humoral immunity and decreases parasite control. Our results show that targeting OX40 can enhance Plasmodium control and that crosstalk between co-inhibitory and co-stimulatory pathways in pathogen-specific CD4 T cells can impact pathogen clearance.


Assuntos
Diferenciação Celular , Imunidade Humoral , Malária/imunologia , Plasmodium/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Receptores OX40/metabolismo , Linfócitos T Auxiliares-Indutores/fisiologia , Animais , Regulação da Expressão Gênica , Humanos , Camundongos
8.
J Biol Chem ; 289(40): 27979-91, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25135637

RESUMO

Cysteine-containing peptides represent an important class of T cell epitopes, yet their prevalence remains underestimated. We have established and interrogated a database of around 70,000 naturally processed MHC-bound peptides and demonstrate that cysteine-containing peptides are presented on the surface of cells in an MHC allomorph-dependent manner and comprise on average 5-10% of the immunopeptidome. A significant proportion of these peptides are oxidatively modified, most commonly through covalent linkage with the antioxidant glutathione. Unlike some of the previously reported cysteine-based modifications, this represents a true physiological alteration of cysteine residues. Furthermore, our results suggest that alterations in the cellular redox state induced by viral infection are communicated to the immune system through the presentation of S-glutathionylated viral peptides, resulting in altered T cell recognition. Our data provide a structural basis for how the glutathione modification alters recognition by virus-specific T cells. Collectively, these results suggest that oxidative stress represents a mechanism for modulating the virus-specific T cell response.


Assuntos
Apresentação de Antígeno , Infecções por Coronavirus/veterinária , Epitopos de Linfócito T/metabolismo , Vírus da Hepatite Murina/imunologia , Doenças dos Roedores/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Cisteína/metabolismo , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Feminino , Glutationa/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/genética , Oxirredução , Doenças dos Roedores/imunologia , Doenças dos Roedores/virologia
9.
Vaccine ; 32(4): 483-91, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24321740

RESUMO

Sporozoite vaccination of both humans and rodents elicits potent anti-malarial immunity, but the dose of sporozoites and the number of immunizations required varies with vaccination approach. Here we examine the immunological basis for superior protection afforded from single-dose vaccination with virulent sporozoites administered under prophylatic chloroquine-cover, referred to as infection-treatment-vaccination (ITV), compared to the well-studied approach of administering radiation-attenuated Plasmodium sporozoites (RAS). Earlier rodent studies utilizing ITV and RAS vaccination suggested a major role of CD8 T cells in reducing liver parasite burden after sporozoite challenge in a BALB/c mouse model. Consistent with this, we find that in C57Bl/6 mice ITV elicits substantially higher parasite-specific CD8 T cell responses than RAS vaccination and enhances immunity against P. yoelii infection. However, we show ITV-induced CD8 T cells are not necessary for protection following liver-stage sporozoite or blood-stage parasite challenge. Mechanistically, we found protection afforded from single-dose ITV is associated with low grade, transient parasitemia shortly following cessation of chloroquine treatment and generation of potent antibody responses to blood-stage parasites. Collectively, our data show the mechanistic basis for enhanced protective immunity against P. yoelli elicited by ITV in highly susceptible C57Bl/6 mice is independent of CD8 T cells. These studies may be relevant in understanding the potent immunity observed with ITV in humans.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Antimaláricas/uso terapêutico , Malária/prevenção & controle , Plasmodium yoelii , Animais , Anticorpos Antiprotozoários/sangue , Antimaláricos/uso terapêutico , Cloroquina/uso terapêutico , Feminino , Malária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia/prevenção & controle , Esporozoítos/imunologia , Vacinas Atenuadas/uso terapêutico
10.
Trends Immunol ; 33(5): 247-54, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22405559

RESUMO

Malaria is caused by complex protozoan Plasmodium parasites that have foiled efforts to develop a protective vaccine. Despite this, it has been known for more than 40 years that immunization with radiation-attenuated, whole Plasmodium sporozoites confers complete protection against malaria challenge. This model gave the rationale for development of recombinant and vectored subunit vaccination strategies that have, however, not yet matched whole sporozoite protective efficacy. Novel attenuation and immunization approaches for whole sporozoite vaccination and a deeper understanding of cellular and humoral protective immune responses that eliminate pre-erythrocytic stages are paving the way for the development of next-generation vaccination strategies that completely prevent malaria.


Assuntos
Malária/prevenção & controle , Plasmodium/imunologia , Vacinação , Animais , Linfócitos T CD8-Positivos/imunologia , Humanos , Malária/imunologia , Vacinas Antimaláricas/uso terapêutico , Plasmodium/crescimento & desenvolvimento , Esporozoítos/imunologia
11.
Cell Host Microbe ; 9(6): 451-62, 2011 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-21669394

RESUMO

While subunit vaccines have shown partial efficacy in clinical trials, radiation-attenuated sporozoites (RAS) remain the "gold standard" for sterilizing protection against Plasmodium infection in human vaccinees. The variability in immunogenicity and replication introduced by the extensive, random DNA damage necessary to generate RAS could be overcome by genetically attenuated parasites (GAP) designed via gene deletion to arrest at defined points during liver-stage development. Here, we demonstrate the principle that late liver stage-arresting GAP induce larger and broader CD8 T cell responses that provide superior protection in inbred and outbred mice compared to RAS or early-arresting GAP immunizations. Late liver stage-arresting GAP also engender high levels of cross-stage and cross-species protection and complete protection when administered by translationally relevant intradermal or subcutaneous routes. Collectively, our results underscore the potential utility of late liver stage-arresting GAP as broadly protective next-generation live-attenuated malaria vaccines and support their potential as a powerful model for identifying antigens to generate cross-stage protection.


Assuntos
Fígado/parasitologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/parasitologia , Deleção de Genes , Humanos , Fígado/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium yoelii/crescimento & desenvolvimento , Esporozoítos/crescimento & desenvolvimento , Esporozoítos/imunologia , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
12.
J Immunol ; 186(10): 5873-84, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21460205

RESUMO

Plasmodium infections are responsible for millions of cases of malaria and ∼1 million deaths annually. Recently, we showed that sterile protection (95%) in BALB/c mice required Plasmodium berghei circumsporozoite protein (CS(252-260))-specific memory CD8 T cells exceeding a threshold of 1% of all PBLs. Importantly, it is not known if Plasmodium species affect the threshold of CS-specific memory CD8 T cells required for protection. Furthermore, C57BL/6 mice immunized with radiation-attenuated parasites are more difficult to protect against Plasmodium sporozoite challenge than similarly immunized BALB/c mice; however, it is not known whether this is the result of different CD8 T cell specificity, functional attributes of CD8 T cells, or mouse strain-specific factors expressed in nonhematopoietic cells. In this article, we show that more CS-specific memory CD8 T cells are required for protection against P. yoelii sporozoite challenge than for protection against P. berghei sporozoite challenge. Furthermore, P. berghei CS(252)-specific CD8 T cells exhibit reduced protection against P. berghei sporozoite challenge in the context of C57BL/6 and C57BL/10 non-MHC-linked genes in CB6F1 and B10.D2 mice, respectively. Generation and immunization of reciprocal chimeric mice between BALB/c and B10.D2 strains revealed that B10 background factors expressed by nonhematopoietic cells increased the threshold required for protection through a CD8 T cell-extrinsic mechanism. Finally, reduced CS-specific memory CD8 T cell protection in P. yoelii-infected BALB/c or P. berghei-infected B10.D2 mice correlated with increased rates of Plasmodium amplification in the liver. Thus, both Plasmodium species and strain-specific background genes in nonhematopoietic cells determine the threshold of memory CD8 T cells required for protection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Plasmodium berghei/imunologia , Plasmodium yoelii/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Animais , Interações Hospedeiro-Parasita , Interferon gama/imunologia , Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA/parasitologia , Plasmodium berghei/efeitos da radiação , Plasmodium yoelii/efeitos da radiação , Subpopulações de Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/imunologia
13.
PLoS Pathog ; 6(7): e1000998, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20657824

RESUMO

Radiation-attenuated Plasmodium sporozoites (RAS) are the only vaccine shown to induce sterilizing protection against malaria in both humans and rodents. Importantly, these "whole-parasite" vaccines are currently under evaluation in human clinical trials. Studies with inbred mice reveal that RAS-induced CD8 T cells targeting liver-stage parasites are critical for protection. However, the paucity of defined T cell epitopes for these parasites has precluded precise understanding of the specific characteristics of RAS-induced protective CD8 T cell responses. Thus, it is not known whether quantitative or qualitative differences in RAS-induced CD8 T cell responses underlie the relative resistance or susceptibility of immune inbred mice to sporozoite challenge. Moreover, whether extraordinarily large CD8 T cell responses are generated and required for protection following RAS immunization, as has been described for CD8 T cell responses following single-antigen subunit vaccination, remains unknown. Here, we used surrogate T cell activation markers to identify and track whole-parasite, RAS-vaccine-induced effector and memory CD8 T cell responses. Our data show that the differential susceptibility of RAS-immune inbred mouse strains to Plasmodium berghei or P. yoelii sporozoite challenge does not result from host- or parasite-specific decreases in the CD8 T cell response. Moreover, the surrogate activation marker approach allowed us for the first time to evaluate CD8 T cell responses and protective immunity following RAS-immunization in outbred hosts. Importantly, we show that compared to a protective subunit vaccine that elicits a CD8 T cell response to a single epitope, diversifying the targeted antigens through whole-parasite RAS immunization only minimally, if at all, reduced the numerical requirements for memory CD8 T cell-mediated protection. Thus, our studies reveal that extremely high frequencies of RAS-induced memory CD8 T cells are required, but may not suffice, for sterilizing anti-Plasmodial immunity. These data provide new insights into protective CD8 T cell responses elicited by RAS-immunization in genetically diverse hosts, information with relevance to developing attenuated whole-parasite vaccines.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunização , Fígado/parasitologia , Vacinas Antimaláricas/imunologia , Esporozoítos/imunologia , Animais , Antimaláricos/imunologia , Imunidade , Memória Imunológica , Fígado/imunologia , Vacinas Antimaláricas/administração & dosagem , Camundongos , Esporozoítos/efeitos da radiação , Vacinas Atenuadas/imunologia
14.
J Immunol ; 184(5): 2528-38, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20097864

RESUMO

Malaria results in >1,000,000 deaths per year worldwide. Although no licensed vaccine exists, much effort is currently focused on subunit vaccines that elicit CD8 T cell responses directed against Plasmodium parasite liver stage Ags. Multiple immune-effector molecules play a role in antimicrobial immunity mediated by memory CD8 T cells, including IFN-gamma, perforin, TRAIL, Fas ligand, and TNF-alpha. However, it is not known which pathways are required for memory CD8 T cell-mediated immunity against liver stage Plasmodium infection. In this study, we used a novel immunization strategy to generate memory CD8 T cells in the BALB/c mouse model of P. berghei or P. yoelii sporozoite infection to examine the role of immune-effector molecules in resistance to the liver stage infection. Our studies reveal that endogenous memory CD8 T cell-mediated protection against both parasite species is, in part, dependent on IFN-gamma, whereas perforin was only critical in protection against P. yoelii. We further show that neutralization of TNF-alpha in immunized mice markedly reduces memory CD8 T cell-mediated protection against both parasite species. Thus, our studies identify IFN-gamma and TNF-alpha as important components of the noncytolytic pathways that underlie memory CD8 T cell-mediated immunity against liver stage Plasmodium infection. Our studies also show that the effector pathways that memory CD8 T cells use to eliminate liver stage infection are, in part, Plasmodium species specific.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium berghei/imunologia , Plasmodium yoelii/imunologia , Esporozoítos/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Culicidae/parasitologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/metabolismo , Feminino , Memória Imunológica/imunologia , Interferon gama/deficiência , Interferon gama/genética , Interferon gama/imunologia , Listeria monocytogenes/imunologia , Hepatopatias Parasitárias/imunologia , Hepatopatias Parasitárias/parasitologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Perforina/deficiência , Perforina/genética , Perforina/imunologia , Transdução de Sinais/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/deficiência , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
15.
PLoS Pathog ; 4(10): e1000186, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18949029

RESUMO

High affinity antigen-specific T cells play a critical role during protective immune responses. Epitope enhancement can elicit more potent T cell responses and can subsequently lead to a stronger memory pool; however, the molecular basis of such enhancement is unclear. We used the consensus peptide-binding motif for the Major Histocompatibility Complex molecule H-2K(b) to design a heteroclitic version of the mouse hepatitis virus-specific subdominant S598 determinant. We demonstrate that a single amino acid substitution at a secondary anchor residue (Q to Y at position 3) increased the stability of the engineered determinant in complex with H-2K(b). The structural basis for this enhanced stability was associated with local alterations in the pMHC conformation as a result of the Q to Y substitution. Recombinant viruses encoding this engineered determinant primed CTL responses that also reacted to the wildtype epitope with significantly higher functional avidity, and protected against selection of virus mutated at a second CTL determinant and consequent disease progression in persistently infected mice. Collectively, our findings provide a basis for the enhanced immunogenicity of an engineered determinant that will serve as a template for guiding the development of heteroclitic T cell determinants with applications in prevention of CTL escape in chronic viral infections as well as in tumor immunity.


Assuntos
Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Epitopos de Linfócito T/genética , Linfócitos T Citotóxicos/imunologia , Animais , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Modulação Antigênica/genética , Epitopos de Linfócito T/química , Antígenos H-2/química , Antígenos H-2/genética , Antígenos H-2/imunologia , Epitopos Imunodominantes/química , Epitopos Imunodominantes/genética , Camundongos , Modelos Biológicos , Modelos Moleculares , Vírus da Hepatite Murina/genética , Vírus da Hepatite Murina/imunologia , Mutagênese Sítio-Dirigida , Estabilidade Proteica , Linfócitos T Citotóxicos/fisiologia , Temperatura , Termodinâmica
16.
J Immunol ; 180(6): 3926-37, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18322201

RESUMO

Cytotoxic T lymphocyte escape occurs in many human infections, as well as mice infected with the JHM strain of mouse hepatitis virus, which exhibit CTL escape variants with mutations in a single epitope from the spike glycoprotein (S510). In all CTL epitopes prone to escape, only a subset of all potential variants is generally detected, even though many of the changes that are not selected would result in evasion of the T cell response. It is postulated that these unselected mutations significantly impair virus fitness. To define more precisely the basis for this preferential selection, we combine x-ray crystallographic studies of the MHC class I (D(b))/S510 complexes with viral reverse genetics to identify a prominent TCR contact residue (tryptophan at position 4) prone to escape mutations. The data show that a mutation that is commonly detected in chronically infected mice (tryptophan to arginine) potently disrupts the topology of the complex, explaining its selection. However, other mutations at this residue, which also abrogate the CTL response, are never selected in vivo even though they do not compromise virus fitness in acutely infected animals or induce a significant de novo CTL response. Thus, while structural analyses of the S510/D(b) complex provide a strong basis for why some CTL escape variants are selected, our results also show that factors other than effects on virus fitness limit the diversification of CD8 T cell epitopes.


Assuntos
Infecções por Coronavirus/imunologia , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/patogenicidade , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Substituição de Aminoácidos/genética , Animais , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Arginina/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Cristalografia por Raios X , Epitopos de Linfócito T/química , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Feminino , Antígenos H-2/química , Antígenos H-2/genética , Antígenos H-2/metabolismo , Antígeno de Histocompatibilidade H-2D , Epitopos Imunodominantes/química , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/genética , Glicoproteína da Espícula de Coronavírus , Linfócitos T Citotóxicos/metabolismo , Triptofano/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
17.
J Immunol ; 177(7): 4426-35, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16982877

RESUMO

The mechanisms regulating IL-4 mRNA stability in differentiated T cells are not known. We found that early exposure of CD4+ T cells to endogenous IL-4 increased IL-4 mRNA stability. This effect of IL-4 was mediated by the RNA-binding protein HuR. IL-4 mRNA interacted with HuR and the dominant binding site was shown within the coding region of IL-4 mRNA. Exposure of CD4+ T cells to IL-4 had no effects on HuR expression or subcellular localization, but triggered HuR binding to IL-4 mRNA. Thus, IL-4 plays a positive role in maintaining IL-4 mRNA stability in CD4+ T cells via a HuR-mediated mechanism.


Assuntos
Antígenos de Superfície/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Regulação da Expressão Gênica/imunologia , Interleucina-4/genética , Estabilidade de RNA/imunologia , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/imunologia , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Imunofluorescência , Expressão Gênica , Immunoblotting , Interleucina-4/metabolismo , Ativação Linfocitária/fisiologia , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Estrutura Secundária de Proteína , RNA Mensageiro/química
18.
Am J Physiol Lung Cell Mol Physiol ; 288(4): L633-40, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15579629

RESUMO

Using a murine model of sepsis, we found that the balance of tissue pro- to anti-inflammatory cytokines directly correlated with severity of infection and mortality. Sepsis was induced in C57BL/6 mice by cecal ligation and puncture (CLP). Liver tissue was analyzed for levels of IL-1beta, IL-1 receptor antagonist (IL-1ra), tumor necrosis factor (TNF)-alpha, and soluble TNF receptor 1 by ELISA. Bacterial DNA was measured using quantitative real-time PCR. After CLP, early predominance of proinflammatory cytokines (6 h) transitioned to anti-inflammatory predominance at 24 h. The elevated anti-inflammatory cytokines were mirrored by increased tissue bacterial levels. The degree of anti-inflammatory response compared with proinflammatory response correlated with the bacterial concentration. To modulate the timing of the anti-inflammatory response, mice were treated with IL-1ra before CLP. This resulted in decreased proinflammatory cytokines, earlier bacterial load, and increased mortality. These studies show that the initial tissue proinflammatory response to sepsis is followed by an anti-inflammatory response. The anti-inflammatory phase is associated with increased bacterial load and mortality. These data suggest that it is the timing and magnitude of the anti-inflammatory response that predicts severity of infection in a murine model of sepsis.


Assuntos
Interleucina-1/metabolismo , Fígado/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Sepse/mortalidade , Sialoglicoproteínas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Ceco/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/genética , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Punções , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sepse/imunologia , Sepse/microbiologia , Sialoglicoproteínas/genética , Fator de Necrose Tumoral alfa/genética
19.
Am J Respir Cell Mol Biol ; 30(6): 844-52, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14742298

RESUMO

Respiratory syncytial virus (RSV) preferentially infects lung epithelial cells. Infected cells remain viable well into the infection. This prolonged survival results from RSV-induced activation of pro-survival pathways, including Akt and extracellular signal-related kinase (ERK). Sphingosine 1-phosphate (S1P) is a sphingolipid metabolite with demonstrated links to cell survival. It is enzymatically generated by sequential activation of ceramidase (generation of sphingosine) and sphingosine kinase (generation of S1P). In these studies, we found that RSV stimulated neutral ceramidase and sphingosine kinase activities in lung epithelial cells. The combined effect of activation of these two enzymes would decrease proapoptotic ceramide and increase antiapoptotic S1P. S1P activated Akt and ERK within minutes, and inhibition of sphingosine kinase blocked RSV-induced ERK and Akt activation, leading to accelerated cell death after viral infection. RSV infection does eventually kill infected cells but activation of cell survival pathways significantly delays cell death. The studies are the first evidence linking sphingolipid metabolites to cell survival mechanisms in the context of a viral infection.


Assuntos
Sobrevivência Celular/fisiologia , Células Epiteliais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Vírus Sinciciais Respiratórios/metabolismo , Amidoidrolases/metabolismo , Animais , Linhagem Celular , Ceramidases , Ativação Enzimática , Células Epiteliais/citologia , Células Epiteliais/virologia , Humanos , Lisofosfolipídeos/metabolismo , Ceramidase Neutra , Proteínas Proto-Oncogênicas c-akt , Mucosa Respiratória/citologia , Infecções por Vírus Respiratório Sincicial , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo
20.
J Immunol ; 171(10): 5107-15, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14607909

RESUMO

A number of lung diseases, including many interstitial lung diseases and HIV infection, are associated with decreases in intracellular thiols. Altered Th1/Th2 T cell balance has also been associated with disease progression in many of the same diseases. IFN-gamma and IL-4 are critical effector cytokines of Th1 and Th2 cells, respectively. To determine the effect of thiols on the production of IFN-gamma and IL-4 by splenocytes, cells were incubated in the presence and the absence of N-acetylcysteine (NAC) and stimulated with alphaCD3 or alphaCD3 and IL-12. Augmenting intracellular soluble thiol pools ( approximately 2-fold) with 15 mM NAC blocked induction of IFN-gamma and increased production of IL-4 without causing significant changes in intracellular glutathione levels. The effect of NAC on IL-4 production was not linked to an increase in STAT6 phosphorylation, as STAT6 levels were decreased, nor did the increase in IL-4 occur with purified CD4 cells. We found that NAC increased splenocyte IL-4 production via an effect on APCs. We also found that NAC increased two IL-4 relevant transcription factors (AP-1) and NFATc. These studies suggest that increasing intracellular reduced thiol pools decreases IL-12 signaling and IFN-gamma production, while increasing IL-4 production. The sum of these effects may contribute to alterations in the balance between Th1 and Th2 responses in lung diseases associated alterations in intracellular thiol pools.


Assuntos
Adjuvantes Imunológicos/fisiologia , Interleucina-4/biossíntese , Líquido Intracelular/fisiologia , Proteínas Nucleares , Compostos de Sulfidrila/fisiologia , Células Th2/imunologia , Células Th2/metabolismo , Acetilcisteína/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Complexo CD3/metabolismo , Complexo CD3/farmacologia , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Células Cultivadas , Citocinas/biossíntese , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Combinação de Medicamentos , Feminino , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Interleucina-12/antagonistas & inibidores , Interleucina-12/farmacologia , Interleucina-4/metabolismo , Interfase/imunologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fatores de Transcrição NFATC , Fator de Transcrição STAT6 , Solubilidade , Baço/citologia , Baço/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/fisiologia , Transativadores/antagonistas & inibidores , Transativadores/metabolismo , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA