Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 201(4): 1186-1193, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29980611

RESUMO

The lung is a complex organ with anatomically distinct pools of T cells that play specific roles in combating infection. Our knowledge regarding the generation and/or maintenance of immunity by parenchymal or circulating T cells has been gathered from either persistent (>60 d) or rapidly cleared (<10 d) infections. However, the roles of these distinct T cell pools in infections that are cleared over the course of several weeks are not understood. Clearance of the highly virulent intracellular bacterium Francisella tularensis subspecies tularensis (Ftt) following pulmonary infection of immune animals is a protracted T cell-dependent process requiring ∼30-40 d and serves as a model for infections that are not acutely controlled. Using this model, we found that intranasal vaccination increased the number of tissue-resident CD4+ effector T cells, and subsequent challenge of immune mice with Ftt led to a significant expansion of polyfunctional parenchymal CD4+ effector T cells compared with the circulating pool. Despite the dominant in vivo response by parenchymal CD4+ T cells after vaccination and challenge, circulating CD4+ T cells were superior at controlling intracellular Ftt replication in vitro. Further examination in vivo revealed temporal requirements for resident and circulating T cells during Ftt infection. These requirements were in direct contrast to other pulmonary infections that are cleared rapidly in immune animals. The data in this study provide important insights into the role of specific T cell populations that will be essential for the design of novel effective vaccines against tularemia and potentially other agents of pulmonary infection.


Assuntos
Vacinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Francisella tularensis/fisiologia , Pulmão/imunologia , Tularemia/imunologia , Animais , Carga Bacteriana , Proliferação de Células , Modelos Animais de Doenças , Feminino , Humanos , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Vacinação
2.
Front Microbiol ; 9: 607, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29670588

RESUMO

Francisella tularensis is a highly infectious bacterial pathogen that causes the potentially fatal disease tularemia. The Live Vaccine Strain (LVS) of F. tularensis subsp. holarctica, while no longer licensed as a vaccine, is used as a model organism for identifying correlates of immunity and bacterial factors that mediate a productive immune response against F. tularensis. Recently, it was reported that two biovars of LVS differed in their virulence and vaccine efficacy. Genetic analysis showed that they differ in ferrous iron homeostasis; lower Fe2+ levels contributed to increased resistance to hydrogen peroxide in the vaccine efficacious LVS biovar. This also correlated with resistance to the bactericidal activity of interferon γ-stimulated murine bone marrow-derived macrophages. We have extended these findings further by showing that a mutant lacking bacterioferritin stimulates poor protection against Schu S4 challenge in a mouse model of tularemia. Together these results suggest that the efficacious biovar of LVS stimulates productive immunity by a mechanism that is dependent on its ability to limit the toxic effects of oxidative stress by maintaining optimally low levels of intracellular Fe2+.

3.
Vaccine ; 35(19): 2575-2581, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28372827

RESUMO

Francisella tularensis subsp. tularensis strain SchuS4 (Ftt) is a highly virulent intracellular bacterium. Inhalation of 10 or fewer organisms results in an acute and potentially lethal disease called pneumonic tularemia. Ftt infections occur naturally in the U.S. and Ftt was developed as a bioweapon. Thus, there is a need for vaccines that protect against this deadly pathogen. Although a live vaccine strain of Francisella tularensis (LVS) exists, LVS fails to generate long-lived protective immunity against modest challenge doses of Ftt. We recently identified an important role for high avidity CD4+ T cells in short-term protection and hypothesized that expanding this pool of cells would improve overall vaccine efficacy with regard to longevity and challenge dose. In support of our hypothesis, application of a prime/boost vaccination strategy increased the pool of high avidity CD4+ T cells which correlated with improved survival following challenge with either increased doses of virulent Ftt or at late time points after vaccination. In summary, we demonstrate that both epitope selection and vaccination strategies that expand antigen-specific T cells correlate with superior immunity to Ftt as measured by survival.


Assuntos
Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Francisella tularensis/imunologia , Pulmão/imunologia , Tularemia/imunologia , Animais , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Esquemas de Imunização , Camundongos Endogâmicos C57BL , Análise de Sobrevida , Tularemia/prevenção & controle , Estados Unidos
4.
J Immunol ; 197(7): 2738-47, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27543611

RESUMO

T cells are the immunological cornerstone in host defense against infections by intracellular bacterial pathogens, such as virulent Francisella tularensis spp. tularensis (Ftt). The general paucity of novel vaccines for Ftt during the past 60 y can, in part, be attributed to the poor understanding of immune parameters required to survive infection. Thus, we developed a strategy utilizing classical immunological tools to elucidate requirements for effective adaptive immune responses directed against Ftt. Following generation of various Francisella strains expressing well-characterized lymphocytic choriomeningitis virus epitopes, we found that survival correlated with persistence of Ag-specific CD4(+) T cells. Function of these cells was confirmed in their ability to more effectively control Ftt replication in vitro. The importance of understanding the Ag-specific response was underscored by our observation that inclusion of an epitope that elicits high-avidity CD4(+) T cells converted a poorly protective vaccine to one that engenders 100% protection. Taken together, these data suggest that improved efficacy of current tularemia vaccine platforms will require targeting appropriate Ag-specific CD4(+) T cell responses and that elucidation of Francisella epitopes that elicit high-avidity CD4(+) T cell responses, specifically in humans, will be required for successful vaccine development.


Assuntos
Vacinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Francisella tularensis/imunologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos
5.
J Immunol ; 196(10): 4227-36, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27029588

RESUMO

A shift in macrophage metabolism from oxidative phosphorylation to aerobic glycolysis is a requirement for activation to effectively combat invading pathogens. Francisella tularensis is a facultative intracellular bacterium that causes an acute, fatal disease called tularemia. Its primary mechanism of virulence is its ability to evade and suppress inflammatory responses while replicating in the cytosol of macrophages. The means by which F. tularensis modulates macrophage activation are not fully elucidated. In this study, we demonstrate that virulent F. tularensis impairs production of inflammatory cytokines in primary macrophages by preventing their shift to aerobic glycolysis, as evidenced by the downregulation of hypoxia inducible factor 1α and failure to upregulate pfkfb3 We also show that Francisella capsule is required for this process. In addition to modulating inflammatory responses, inhibition of glycolysis in host cells is also required for early replication of virulent Francisella Taken together, our data demonstrate that metabolic reprogramming of host cells by F. tularensis is a key component of both inhibition of host defense mechanisms and replication of the bacterium.


Assuntos
Cápsulas Bacterianas/imunologia , Reprogramação Celular , Francisella tularensis/patogenicidade , Inflamação/imunologia , Macrófagos/imunologia , Animais , Citocinas/imunologia , Regulação para Baixo , Glicólise , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfofrutoquinase-2/metabolismo , Tularemia/imunologia , Virulência
6.
Clin Vaccine Immunol ; 22(1): 119-28, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25410207

RESUMO

Francisella tularensis is an intracellular, Gram-negative bacterium that causes the fatal disease tularemia. Currently, there are no licensed vaccines for tularemia and the requirements for protection against infection are poorly defined. To identify correlates of vaccine-induced immunity against tularemia, we compared different strains of the live vaccine strain (LVS) for their relative levels of virulence and ability to protect C57BL/6 mice against challenge with virulent F. tularensis strain SchuS4. Successful vaccination, as defined by survival of C57BL/6 mice, was correlated with significantly greater numbers of effector T cells in the spleen and lung. Further, lung cells and splenocytes from fully protected animals were more effective than lung cells and splenocytes from vaccinated but nonimmune animals in limiting intracellular replication of SchuS4 in vitro. Together, our data provide a unique model to compare efficacious vaccines to nonefficacious vaccines, which will enable comprehensive identification of host and bacterial components required for immunization against tularemia.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Linfócitos T/imunologia , Tularemia/imunologia , Tularemia/prevenção & controle , Animais , Vacinas Bacterianas/administração & dosagem , Modelos Animais de Doenças , Feminino , Francisella tularensis/crescimento & desenvolvimento , Camundongos Endogâmicos C57BL , Baço/imunologia , Análise de Sobrevida
7.
Front Microbiol ; 5: 438, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25191316

RESUMO

Activation of the inflammasome is important for the detection and clearance of cytosolic pathogens. In contrast to avirulent Francisella novicida (Fn), infection with virulent Francisella tularensis ssp tularensis does not trigger activation of the host AIM2 inflammasome. Here we show that differential activation of AIM2 following Francisella infection is due to sensitivity of each isolate to reactive oxygen species (ROS). ROS present at the outset of Fn infection contributes to activation of the AIM2 inflammasome, independent of NLRP3 and NADPH oxidase. Rather, mitochondrial ROS (mROS) is critical for Fn stimulation of the inflammasome. This study represents the first demonstration of the importance of mROS in the activation of the AIM2 inflammasome by bacteria. Our results also demonstrate that bacterial resistance to mROS is a mechanism of virulence for early evasion of detection by the host.

8.
PLoS One ; 8(12): e82096, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324751

RESUMO

Virulent Francisella tularensis ssp tularensis is an intracellular, Gram negative bacterium that causes acute lethal disease following inhalation of fewer than 15 organisms. Pathogenicity of Francisella infections is tied to its unique ability to evade and suppress inflammatory responses in host cells. It has been proposed that induction of alternative activation of infected macrophages is a mechanism by which attenuated Francisella species modulate host responses. In this report we reveal that neither attenuated F. tularensis Live Vaccine Strain (LVS) nor virulent F. tularensis strain SchuS4 induce alternative activation of macrophages in vitro or in vivo. LVS, but not SchuS4, provoked production of arginase1 independent of alternative activation in vitro and in vivo. However, absence of arginase1 did not significantly impact intracellular replication of LVS or SchuS4. Together our data establish that neither induction of alternative activation nor expression of arginase1 are critical features of disease mediated by attenuated or virulent Francisella species.


Assuntos
Arginase/biossíntese , Francisella tularensis/fisiologia , Ativação de Macrófagos , Macrófagos/enzimologia , Macrófagos/microbiologia , Animais , Citocinas/metabolismo , Indução Enzimática , Francisella tularensis/crescimento & desenvolvimento , Francisella tularensis/patogenicidade , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Tularemia/imunologia , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinas Atenuadas/imunologia
9.
Clin Vaccine Immunol ; 20(10): 1531-40, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23925884

RESUMO

Francisella tularensis is a Gram-negative facultative intracellular pathogen that causes an acute lethal respiratory disease in humans. The heightened virulence of the pathogen is linked to its unique ability to inhibit Toll-like receptor (TLR)-mediated inflammatory responses. The bacterial component and mechanism of this inhibition are unknown. Here we show that lipids isolated from virulent but not attenuated strains of F. tularensis are not detected by host cells, inhibit production of proinflammatory cytokines by primary macrophages in response to known TLR ligands, and suppress neutrophil recruitment in vivo. We further show that lipid-mediated inhibition of inflammation is dependent on TLR2, MyD88, and the nuclear hormone and fatty acid receptor peroxisome proliferator-activated receptor α (PPARα). Pathogen lipid-mediated interference with inflammatory responses through the engagement of TLR2 and PPARα represents a novel manipulation of host signaling pathways consistent with the ability of highly virulent F. tularensis to efficiently evade host immune responses.


Assuntos
Francisella tularensis/imunologia , Imunossupressores/imunologia , Lipídeos/imunologia , PPAR alfa/antagonistas & inibidores , Pneumonia Bacteriana/imunologia , Receptor 2 Toll-Like/antagonistas & inibidores , Animais , Citocinas/metabolismo , Imunossupressores/isolamento & purificação , Lipídeos/isolamento & purificação , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , PPAR alfa/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/prevenção & controle , Receptor 2 Toll-Like/imunologia
10.
J Immunol ; 190(6): 2756-66, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23378429

RESUMO

B1a cells are an important source of natural Abs, Abs directed against T-independent Ags, and are a primary source of IL-10. Bruton's tyrosine kinase (btk) is a cytoplasmic kinase that is essential for mediating signals from the BCR and is critical for development of B1a cells. Consequentially, animals lacking btk have few B1a cells, minimal Ab responses, and can preferentially generate Th1-type immune responses following infection. B1a cells have been shown to aid in protection against infection with attenuated Francisella tularensis, but their role in infection mediated by fully virulent F. tularensis is not known. Therefore, we used mice with defective btk (CBA/CaHN-Btk(XID)/J [XID mice]) to determine the contribution of B1a cells in defense against the virulent F. tularensis ssp. tularensis strain SchuS4. Surprisingly, XID mice displayed increased resistance to pulmonary infection with F. tularensis. Specifically, XID mice had enhanced clearance of bacteria from the lung and spleen and significantly greater survival of infection compared with wild-type controls. We revealed that resistance to infection in XID mice was associated with decreased numbers of IL-10-producing B1a cells and concomitant increased numbers of IL-12-producing macrophages and IFN-γ-producing NK/NKT cells. Adoptive transfer of wild-type B1a cells into XID mice reversed the control of bacterial replication. Similarly, depletion of NK/NKT cells also increased bacterial burdens in XID mice. Together, our data suggest B cell-NK/NKT cell cross-talk is a critical pivot controlling survival of infection with virulent F. tularensis.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/microbiologia , Francisella tularensis/imunologia , Predisposição Genética para Doença , Células Matadoras Naturais/imunologia , Células T Matadoras Naturais/imunologia , Animais , Anticorpos Antibacterianos/biossíntese , Subpopulações de Linfócitos B/transplante , Comunicação Celular/genética , Comunicação Celular/imunologia , Células Cultivadas , Exsudatos e Transudatos/microbiologia , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Células Matadoras Naturais/microbiologia , Células Matadoras Naturais/patologia , Camundongos , Camundongos Endogâmicos CBA , Camundongos Mutantes , Células T Matadoras Naturais/microbiologia , Células T Matadoras Naturais/patologia , Peritônio/imunologia , Peritônio/microbiologia , Peritônio/patologia , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia , Análise de Sobrevida , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/microbiologia , Tularemia/imunologia , Tularemia/mortalidade , Tularemia/patologia , Virulência/genética , Virulência/imunologia
11.
PLoS One ; 7(5): e37752, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22662210

RESUMO

Tularemia, caused by the gram-negative bacterium Francisella tularensis, is a severe, sometimes fatal disease. Interest in tularemia has increased over the last decade due to its history as a biological weapon. In particular, development of novel vaccines directed at protecting against pneumonic tularemia has been an important goal. Previous work has demonstrated that, when delivered at very high inoculums, administration of live, highly attenuated strains of virulent F. tularensis can protect against tularemia. However, lower vaccinating inoculums did not offer similar immunity. One concern of using live vaccines is that the host may develop mild tularemia in response to infection and use of high inoculums may contribute to this issue. Thus, generation of a live vaccine that can efficiently protect against tularemia when delivered in low numbers, e.g. <100 organisms, may address this concern. Herein we describe the ability of three defined, attenuated mutants of F. tularensis SchuS4, deleted for FTT0369c, FTT1676, or FTT0369c and FTT1676, respectively, to engender protective immunity against tularemia when delivered at concentrations of approximately 50 or fewer bacteria. Attenuated strains for use as vaccines were selected by their inability to efficiently replicate in macrophages in vitro and impaired replication and dissemination in vivo. Although all strains were defective for replication in vitro within macrophages, protective efficacy of each attenuated mutant was correlated with their ability to modestly replicate and disseminate in the host. Finally, we demonstrate the parenteral vaccination with these strains offered superior protection against pneumonic tularemia than intranasal vaccination. Together our data provides proof of principle that low dose attenuated vaccines may be a viable goal in development of novel vaccines directed against tularemia.


Assuntos
Vacinas Bacterianas/administração & dosagem , Francisella tularensis/imunologia , Tularemia/prevenção & controle , Animais , Vacinas Bacterianas/imunologia , Feminino , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação , Tularemia/mortalidade , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Virulência/genética
12.
PLoS One ; 7(3): e33349, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22428026

RESUMO

Francisella tularensis is a facultative intracellular bacterium and the causative agent of tularemia. Development of novel vaccines and therapeutics for tularemia has been hampered by the lack of understanding of which immune components are required to survive infection. Defining these requirements for protection against virulent F. tularensis, such as strain SchuS4, has been difficult since experimentally infected animals typically die within 5 days after exposure to as few as 10 bacteria. Such a short mean time to death typically precludes development, and therefore assessment, of immune responses directed against virulent F. tularensis. To enable identification of the components of the immune system that are required for survival of virulent F. tularensis, we developed a convalescent model of tularemia in C57Bl/6 mice using low dose antibiotic therapy in which the host immune response is ultimately responsible for clearance of the bacterium. Using this model we demonstrate αßTCR(+) cells, γδTCR(+) cells, and B cells are necessary to survive primary SchuS4 infection. Analysis of mice deficient in specific soluble mediators shows that IL-12p40 and IL-12p35 are essential for survival of SchuS4 infection. We also show that IFN-γ is required for survival of SchuS4 infection since mice lacking IFN-γR succumb to disease during the course of antibiotic therapy. Finally, we found that both CD4(+) and CD8(+) cells are the primary producers of IFN-γand that γδTCR(+) cells and NK cells make a minimal contribution toward production of this cytokine throughout infection. Together these data provide a novel model that identifies key cells and cytokines required for survival or exacerbation of infection with virulent F. tularensis and provides evidence that this model will be a useful tool for better understanding the dynamics of tularemia infection.


Assuntos
Linfócitos B/imunologia , Modelos Animais de Doenças , Imunidade Celular/imunologia , Linfócitos T/imunologia , Tularemia/imunologia , Análise de Variância , Animais , Antibacterianos/uso terapêutico , Citocinas/metabolismo , Citometria de Fluxo , Interferon gama/imunologia , Interleucina-12/imunologia , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/metabolismo , Tularemia/tratamento farmacológico
13.
Vaccine ; 28(40): 6562-72, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20688042

RESUMO

Protection against the intracellular bacterium Francisella tularensis within weeks of vaccination is thought to involve both cellular and humoral immune responses. However, the relative roles for cellular and humoral immunity in long lived protection against virulent F. tularensis are not well established. Here, we dissected the correlates of immunity to pulmonary infection with virulent F. tularensis strain SchuS4 in mice challenged 30 and 90 days after subcutaneous vaccination with LVS. Regardless of the time of challenge, LVS vaccination protected approximately 90% of SchuS4 infected animals. Surprisingly, control of bacterial replication in the lung during the first 7 days of infection was not required for survival of SchuS4 infection in vaccinated mice. Control and survival of virulent F. tularensis strain SchuS4 infection within 30 days of vaccination was associated with high titers of SchuS4 agglutinating antibodies, and IFN-γ production by multiple cell types in both the lung and spleen. In contrast, survival of SchuS4 infection 90 days after vaccination was correlated only with IFN-γ producing splenocytes and activated T cells in the spleen. Together these data demonstrate that functional agglutinating antibodies and strong mucosal immunity are correlated with early control of pulmonary infections with virulent F. tularensis. However, early mucosal immunity may not be required to survive F. tularensis infection. Instead, survival of SchuS4 infection at extended time points after immunization was only associated with production of IFN-γ and activation of T cells in peripheral organs.


Assuntos
Vacinas Bacterianas/imunologia , Imunidade Celular , Pneumopatias/microbiologia , Tularemia/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Feminino , Francisella tularensis/imunologia , Imunidade Humoral , Interferon gama/imunologia , Pulmão/citologia , Pulmão/imunologia , Pneumopatias/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Organismos Livres de Patógenos Específicos , Baço/citologia , Baço/imunologia , Linfócitos T/imunologia , Tularemia/imunologia
14.
J Immunol ; 183(7): 4593-600, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19752236

RESUMO

Opsonization by Abs represents a critical component of the host immune response against many pathogens. The mechanisms by which virulent microbes evade this protective response are not completely understood. In disease mediated by Francisella tularensis, Ab can effectively protect against infections with attenuated strains, for example, LVS, but not virulent strains such as SchuS4. Thus, it is likely that SchuS4 has mechanisms, which are not present in LVS, that allow evasion of opsonization by Ab, dampening the protective effects of these host molecules. Here we demonstrate that evasion of Ab-mediated opsonization and phagocytosis by the highly virulent SchuS4 is associated with its ability to bind the host serine protease plasmin. SchuS4, but not the closely related LVS, bound active plasmin. Plasmin bound SchuS4 degraded exogenous and opsonizing Abs, whereas LVS failed to do so. Furthermore, plasmin-mediated inhibition of Ab opsonization by SchuS4 also inhibited Ab-mediated uptake of this bacterium by macrophages. Ab-mediated uptake of uncoated and opsonized SchuS4 elicited a strong proinflammatory response in infected macrophages. However, plasmin-coated, opsonized SchuS4 poorly elicited production of these protective proinflammatory cytokines. This unique host-pathogen interplay is a novel immune evasion strategy utilized by virulent F. tularensis, and it provides one explanation for the ability of Ab to protect against attenuated, but not virulent, strains of F. tularensis. This mechanism may also represent a more common hereto unrecognized strategy by which virulent bacteria evade detection and clearance by Ig.


Assuntos
Anticorpos Antibacterianos/metabolismo , Fibrinolisina/fisiologia , Francisella tularensis/imunologia , Francisella tularensis/patogenicidade , Proteínas Opsonizantes/metabolismo , Animais , Aderência Bacteriana/imunologia , Células Cultivadas , Fibrinolisina/metabolismo , Francisella tularensis/metabolismo , Humanos , Camundongos , Fagocitose/imunologia , Plasminogênio/metabolismo , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/metabolismo , Virulência/imunologia
15.
J Immunol ; 182(12): 8063-70, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494332

RESUMO

A vaccine is likely the most effective strategy for controlling human chlamydial infections. Recent studies have shown immunization with Chlamydia muridarum major outer membrane protein (MOMP) can induce significant protection against infection and disease in mice if its native trimeric structure is preserved (nMOMP). The objective of this study was to investigate the immunogenicity and vaccine efficacy of Chlamydia trachomatis nMOMP in a nonhuman primate trachoma model. Cynomolgus monkeys (Macaca fascicularis) were immunized systemically with nMOMP, and monkeys were challenged ocularly. Immunization induced high serum IgG and IgA ELISA Ab titers, with Abs displaying high strain-specific neutralizing activity. The PBMCs of immunized monkeys produced a broadly cross-reactive, Ag-specific IFN-gamma response equivalent to that induced by experimental infection. Immunized monkeys exhibited a significant decrease in infectious burden during the early peak shedding periods (days 3-14). However, at later time points, they exhibited no difference from control animals in either burden or duration of infection. Immunization had no effect on the progression of ocular disease. These results show that systemically administered nMOMP is highly immunogenic in nonhuman primates and elicits partially protective immunity against ocular chlamydial challenge. This is the first time a subunit vaccine has shown a significant reduction in ocular shedding in nonhuman primates. A partially protective vaccine, particularly one that reduces infectious burden after primary infection of children, could interrupt the natural trachoma reinfection cycle. This would have a beneficial effect on the transmission between children and sensitized adults which drives blinding inflammatory disease.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/imunologia , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/prevenção & controle , Chlamydia trachomatis/imunologia , Macaca fascicularis/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Especificidade de Anticorpos , Infecções por Chlamydia/patologia , Infecções por Chlamydia/transmissão , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Células HeLa , Humanos , Cinética , Leucócitos/imunologia , Leucócitos/metabolismo , Masculino , Desnaturação Proteica , Titulometria
16.
Cell Microbiol ; 11(7): 1128-50, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19388904

RESUMO

Summary The highly infectious bacterium Francisella tularensis is a facultative intracellular pathogen, whose virulence requires proliferation inside host cells, including macrophages. Here we have performed a global transcriptional profiling of the highly virulent F. tularensis ssp. tularensis Schu S4 strain during its intracellular cycle within primary murine macrophages, to characterize its intracellular biology and identify pathogenic determinants based on their intracellular expression profiles. Phagocytosed bacteria rapidly responded to their intracellular environment and subsequently altered their transcriptional profile. Differential gene expression profiles were revealed that correlated with specific intracellular locale of the bacteria. Upregulation of general and oxidative stress response genes was a hallmark of the early phagosomal and late endosomal stages, while induction of transport and metabolic genes characterized the cytosolic replication stage. Expression of the Francisella Pathogenicity Island (FPI) genes, which are required for intracellular proliferation, increased during the intracellular cycle. Similarly, 27 chromosomal loci encoding putative hypothetical, secreted, outer membrane proteins or transcriptional regulators were identified as upregulated. Among these, deletion of FTT0383, FTT0369c or FTT1676 abolished the ability of Schu S4 to survive or proliferate intracellularly and cause lethality in mice, therefore identifying novel determinants of Francisella virulence from their intracellular expression profile.


Assuntos
Francisella tularensis/fisiologia , Perfilação da Expressão Gênica , Macrófagos/microbiologia , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Virulência/biossíntese , Animais , Transporte Biológico , Células Cultivadas , Citosol/microbiologia , Endossomos/microbiologia , Francisella tularensis/crescimento & desenvolvimento , Francisella tularensis/patogenicidade , Genes Bacterianos , Ilhas Genômicas , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Estresse Oxidativo , Fagossomos/microbiologia , Estresse Fisiológico , Virulência
17.
Infect Immun ; 76(6): 2273-83, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18347045

RESUMO

Chlamydia trachomatis possesses a cryptic 7.5-kb plasmid of unknown function. Here, we describe a comprehensive molecular and biological characterization of the naturally occurring plasmidless human C. trachomatis strain L2(25667R). We found that despite minimal chromosomal polymorphisms, the LGV strain L2(25667R) was indistinguishable from plasmid-positive strain L2(434) with regard to its in vitro infectivity characteristics such as growth kinetics, plaquing efficiency, and plaque size. The only in vitro phenotypic differences between L2(434) and L2(25667R) were the accumulation of glycogen granules in the inclusion matrix and the lack of the typical intrainclusion Brownian-like movement characteristic of C. trachomatis strains. Conversely, we observed a marked difference between the two strains in their abilities to colonize and infect the female mouse genital tract. The 50% infective dose of plasmidless strain L2(25667R) was 400-fold greater (4 x 10(6) inclusion-forming units [IFU]) than that of plasmid-bearing strain L2(434) (1 x 10(4) IFU). Transcriptome analysis of the two strains demonstrated a decrease in the transcript levels of a subset of chromosomal genes for strain L2(25667R). Among those genes was glgA, encoding glycogen synthase, a finding consistent with the failure of L2(25667R) to accumulate glycogen granules. These findings support a primary role for the plasmid in in vivo infectivity and suggest that virulence is controlled, at least in part, by the plasmid's ability to regulate the expression of chromosomal genes. Our findings have important implications in understanding a role for the plasmid in the pathogenesis of human infection and disease.


Assuntos
Chlamydia trachomatis/fisiologia , Cromossomos Bacterianos/genética , Plasmídeos/fisiologia , Transcrição Gênica/fisiologia , Fatores de Virulência/fisiologia , Animais , Técnicas Bacteriológicas , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/classificação , Chlamydia trachomatis/citologia , Chlamydia trachomatis/genética , Feminino , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Plasmídeos/genética , Polimorfismo Genético , Análise Serial de Proteínas , Vaginose Bacteriana/microbiologia , Fatores de Virulência/genética
18.
J Infect Dis ; 197(3): 449-56, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18199030

RESUMO

Chlamydia trachomatis is the etiological agent of trachoma, the leading cause of preventable blindness. Trachoma presents distinct clinical syndromes ranging from mild and self-limiting to severe inflammatory disease. The underlying host and pathogen factors responsible for these diverse clinical outcomes are unclear. To assess the role played by pathogen variation in disease outcome, we analyzed the genomes of 4 trachoma strains representative of the 3 major trachoma serotypes, using microarray-based comparative genome sequencing. Outside of ompA, trachoma strains differed primarily in a very small subset of genes (n = 22). These subtle genetic variations were manifested in profound differences in virulence as measured by in vitro growth rate, burst size, plaque morphology, and interferon-gamma sensitivity but most importantly in virulence as shown by ocular infection of nonhuman primates. Our findings are the first to identify genes that correlate with differences in pathogenicity among trachoma strains.


Assuntos
Chlamydia trachomatis/genética , Variação Genética , Genoma Bacteriano , Primatas/microbiologia , Animais , Chlamydia trachomatis/isolamento & purificação , Chlamydia trachomatis/patogenicidade , Células HeLa , Humanos , Macaca fascicularis , Masculino , Polimorfismo Genético , Tracoma/microbiologia
19.
Infect Immun ; 75(12): 5669-77, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17908813

RESUMO

Chlamydia trachomatis is an obligate intracellular pathogen of humans that exhibits species-specific biological characteristics in its early interactions with host cells that are likely important to pathogenesis. One such characteristic is the tyrosine phosphorylation (Tyr-P) of an approximately 70-kDa polypeptide that occurs only after infection of mammalian cells by human strains. We sought to identify this protein because of its potential significance to the pathogenesis of human chlamydial infections. Using an immunoproteomic approach we identified the host protein ezrin, a member of the ezrin-radixin-moesin (ERM) protein family that serves as a physical link between host cell receptors and the actin cytoskeleton. Confocal microscopy studies showed colocalization of ezrin and actin at the tips and crypts of microvilli, the site of chlamydial attachment and entry, respectively. To demonstrate a functional role for ezrin we infected cells with a dominant-negative (DN) ezrin phenotype or treated cells with ezrin-specific small interfering RNA (siRNA). We found that both DN and siRNA-treated cells were significantly less susceptible to infection by human chlamydial strains. Moreover, we demonstrated that inhibition of infection in ezrin DN cells occurred at the stage of chlamydial entry. We hypothesize that the C. trachomatis-specific Tyr-P of ezrin might relate to an undefined species-specific mechanism of pathogen entry that involves chlamydial specific ligand(s) and host cell coreceptor usage.


Assuntos
Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/patogenicidade , Proteínas do Citoesqueleto/metabolismo , Transporte Proteico/fisiologia , Tirosina/metabolismo , Actinas/metabolismo , Actinas/fisiologia , Animais , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/metabolismo , Proteínas do Citoesqueleto/fisiologia , Citoesqueleto/metabolismo , Endocitose/fisiologia , Cobaias , Células HeLa , Humanos , Fosforilação , Transdução de Sinais , Especificidade da Espécie , Suínos
20.
Cell Microbiol ; 9(9): 2289-98, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17501981

RESUMO

Chlamydia trachomatis and C. muridarum, human and mouse pathogens, respectively, share more than 99% of open reading frames (ORFs) but differ in a cytotoxin locus. Presence or absence of cytotoxin gene(s) in these strains correlates with their ability to grow in IFN-gamma treated mouse cells. Growth of toxin-positive C. muridarum is not affected in IFN-gamma treated cells, whereas growth of toxin-negative C. trachomatis is inhibited. We previously reported that this difference in IFN-gamma sensitivity is important to the in vivo infection tropism of these pathogens. Here we describe a phenotypic rescue assay that utilizes C. muridarum gamma irradiated killed elementary bodies (iEB) to rescue C. trachomatis infectivity in IFN-gamma treated mouse cells. Rescue by iEB was temporal, maximal early post infection, directly related to multiplicity of iEB infection, and was independent of de novo chlamydial transcription. Lastly, C. muridarum iEB vacuoles and C. trachomatis inclusions were not fusogenic, suggesting the factor(s) responsible for rescue was secreted or exposed to the cytosol where it inactivated IFN-gamma induced effectors. Chlamydial phenotypic rescue may have broader utility for the study of other EB associated virulence factors that function early in the interaction of chlamydiae with host cells.


Assuntos
Chlamydia muridarum/fisiologia , Chlamydia trachomatis/fisiologia , Interferon gama/metabolismo , Animais , Chlamydia muridarum/citologia , Chlamydia muridarum/genética , Chlamydia muridarum/efeitos da radiação , Chlamydia trachomatis/citologia , Chlamydia trachomatis/genética , Raios gama , Genoma Bacteriano , Células HeLa , Humanos , Camundongos , Fenótipo , Vacúolos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...