Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
3.
Infect Immun ; 87(5)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30804102

RESUMO

Plague is a rapidly lethal human disease caused by the bacterium Yersinia pestis This study demonstrated that the Y. pestis plasminogen activator Pla, a protease that promotes fibrin degradation, thwarts T cell-mediated defense against fully virulent Y. pestis Introducing a single point mutation into the active site of Pla suffices to render fully virulent Y. pestis susceptible to primed T cells. Mechanistic studies revealed essential roles for fibrin during T cell-mediated defense against Pla-mutant Y. pestis Moreover, the efficacy of T cell-mediated protection against various Y. pestis strains displayed an inverse relationship with their levels of Pla activity. Together, these data indicate that Pla functions to thwart fibrin-dependent T cell-mediated defense against plague. Other important human bacterial pathogens, including staphylococci, streptococci, and borrelia, likewise produce virulence factors that promote fibrin degradation. The discovery that Y. pestis thwarts T cell defense by promoting fibrinolysis suggests novel therapeutic approaches to amplifying T cell responses against human pathogens.


Assuntos
Fibrinólise/imunologia , Peste/imunologia , Ativadores de Plasminogênio/imunologia , Linfócitos T/imunologia , Fatores de Virulência/imunologia , Yersinia pestis/patogenicidade , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL
4.
Oncotarget ; 7(23): 33581-94, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-27177221

RESUMO

Influenza and pneumonia are leading causes of death in elderly populations. With age, there is an increased inflammatory response and slower viral clearance during influenza infection which increases the risk of extended illness and mortality. Here we employ a preclinical murine model of influenza infection to examine the protective capacity of vaccination with influenza nucleoprotein (NP). While NP vaccination reduces influenza-induced lung inflammation in young mice, aged mice do not show this reduction, but are protected from influenza-induced mortality. Aged mice do make a significant amount of NP-specific IgG and adoptive transfer experiments show that NP antibody can protect from death but cannot reduce lung inflammation. Furthermore, young but not aged vaccinated mice generate significant numbers of NP-specific T cells following subsequent infection and few of these T cells are found in aged lungs early during infection. Importantly, aged CD4 T cells have a propensity to differentiate towards a T follicular helper (Tfh) phenotype rather than a T helper 1 (Th1) phenotype that predominates in the young. Since Th1 cells are important in viral clearance, reduced Th1 differentiation in the aged is critical and could account for some or all of the age-related differences in vaccine responses and infection resolution.


Assuntos
Envelhecimento/imunologia , Diferenciação Celular/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Nucleocapsídeo , Proteínas de Ligação a RNA/imunologia , Proteínas do Core Viral/imunologia
5.
PLoS One ; 11(4): e0152968, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27046148

RESUMO

Sepsis, a systemic inflammatory response to infection, is often accompanied by abnormalities of blood coagulation. Prior work with a mouse model of sepsis induced by cecal ligation and puncture (CLP) suggested that the protease factor XIa contributed to disseminated intravascular coagulation (DIC) and to the cytokine response during sepsis. We investigated the importance of factor XI to cytokine and coagulation responses during the first 24 hours after CLP. Compared to wild type littermates, factor XI-deficient (FXI-/-) mice had a survival advantage after CLP, with smaller increases in plasma levels of TNF-α and IL-10 and delayed IL-1ß and IL-6 responses. Plasma levels of serum amyloid P, an acute phase protein, were increased in wild type mice 24 hours post-CLP, but not in FXI-/- mice, supporting the impression of a reduced inflammatory response in the absence of factor XI. Surprisingly, there was little evidence of DIC in mice of either genotype. Plasma levels of the contact factors factor XII and prekallikrein were reduced in WT mice after CLP, consistent with induction of contact activation. However, factor XII and PK levels were not reduced in FXI-/- animals, indicating factor XI deficiency blunted contact activation. Intravenous infusion of polyphosphate into WT mice also induced changes in factor XII, but had much less effect in FXI deficient mice. In vitro analysis revealed that factor XIa activates factor XII, and that this reaction is enhanced by polyanions such polyphosphate and nucleic acids. These data suggest that factor XI deficiency confers a survival advantage in the CLP sepsis model by altering the cytokine response to infection and blunting activation of the contact (kallikrein-kinin) system. The findings support the hypothesis that factor XI functions as a bidirectional interface between contact activation and thrombin generation, allowing the two processes to influence each other.


Assuntos
Coinfecção/metabolismo , Citocinas/metabolismo , Deficiência do Fator XI/metabolismo , Peptídeo Hidrolases/metabolismo , Sepse/metabolismo , Animais , Coinfecção/enzimologia , Ativação Enzimática , Camundongos , Sepse/enzimologia
6.
Clin Infect Dis ; 60(2): 292-7, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25273081

RESUMO

Combination antiretroviral therapy can suppress human immunodeficiency virus (HIV) infection but cannot completely eradicate the virus. A major obstacle in the quest for a cure is the difficulty in targeting and measuring latently infected cells. To date, a single person seems to have been cured of HIV. Hematopoietic stem cell transplantation (HSCT) preceded this cancer patient's long-term sustained HIV remission, but researchers have been unable to replicate this cure, and the mechanisms that led to HIV remission remain to be established. In February 2014, the National Institute of Allergy and Infectious Diseases sponsored a workshop that provided a venue for in-depth discussion of whether HSCT could be exploited to cure HIV in cancer patients requiring such procedures. Participants also discussed how HSCT might be applied to a broader community of HIV-infected persons in whom the risks of HSCT currently outweigh the likelihood and benefits of HIV cure.


Assuntos
Infecções por HIV/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Pesquisa Biomédica/tendências , Humanos
7.
PLoS Pathog ; 10(5): e1004142, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24854422

RESUMO

Septic pneumonias resulting from bacterial infections of the lung are a leading cause of human death worldwide. Little is known about the capacity of CD8 T cell-mediated immunity to combat these infections and the types of effector functions that may be most effective. Pneumonic plague is an acutely lethal septic pneumonia caused by the Gram-negative bacterium Yersinia pestis. We recently identified a dominant and protective Y. pestis antigen, YopE69-77, recognized by CD8 T cells in C57BL/6 mice. Here, we use gene-deficient mice, Ab-mediated depletion, cell transfers, and bone marrow chimeric mice to investigate the effector functions of YopE69-77-specific CD8 T cells and their relative contributions during pulmonary Y. pestis infection. We demonstrate that YopE69-77-specific CD8 T cells exhibit perforin-dependent cytotoxicity in vivo; however, perforin is dispensable for YopE69-77-mediated protection. In contrast, YopE69-77-mediated protection is severely impaired when production of TNFα and IFNγ by CD8 T cells is simultaneously ablated. Interestingly, TNFα is absolutely required at the time of challenge infection and can be provided by either T cells or non-T cells, whereas IFNγ provided by T cells prior to challenge appears to facilitate the differentiation of optimally protective CD8 T cells. We conclude that cytokine production, not cytotoxicity, is essential for CD8 T cell-mediated control of pulmonary Y. pestis infection and we suggest that assays detecting Ag-specific TNFα production in addition to antibody titers may be useful correlates of vaccine efficacy against plague and other acutely lethal septic bacterial pneumonias.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade Celular/genética , Interferon gama/fisiologia , Peste/imunologia , Pneumonia Bacteriana/imunologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Yersinia pestis/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peste/complicações , Peste/genética , Pneumonia Bacteriana/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Fator de Necrose Tumoral alfa/genética
8.
Infect Immun ; 81(6): 2123-32, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23545300

RESUMO

Immunomodulatory agents potentially represent a new class of broad-spectrum antimicrobials. Here, we demonstrate that prophylaxis with immunomodulatory cytosine-phosphate-guanidine (CpG) oligodeoxynucleotide (ODN), a toll-like receptor 9 (TLR9) agonist, confers protection against Yersinia pestis, the etiologic agent of plague. The data establish that intranasal administration of CpG ODN 1 day prior to lethal pulmonary exposure to Y. pestis strain KIM D27 significantly improves survival of C57BL/6 mice and reduces bacterial growth in hepatic tissue, despite paradoxically increasing bacterial growth in the lung. All of these CpG ODN-mediated impacts, including the increased pulmonary burden, are TLR9 dependent, as they are not observed in TLR9-deficient mice. The capacity of prophylactic intranasal CpG ODN to enhance survival does not require adaptive immunity, as it is evident in mice lacking B and/or T cells; however, the presence of T cells improves long-term survival. The prophylactic regimen also improves survival and reduces hepatic bacterial burden in mice challenged intraperitoneally with KIM D27, indicating that intranasal delivery of CpG ODN has systemic impacts. Indeed, intranasal prophylaxis with CpG ODN provides significant protection against subcutaneous challenge with Y. pestis strain CO92 even though it fails to protect mice from intranasal challenge with that fully virulent strain.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Oligodesoxirribonucleotídeos/administração & dosagem , Peste/prevenção & controle , Yersinia pestis , Adjuvantes Imunológicos/farmacologia , Administração Intranasal , Animais , Regulação da Expressão Gênica/imunologia , Fígado/microbiologia , Pulmão/citologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/farmacologia , Organismos Livres de Patógenos Específicos , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Virulência , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
9.
J Immunol ; 190(8): 4149-61, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23487423

RESUMO

The Gram-negative bacterium Yersinia pestis causes plague, a rapidly progressing and often fatal disease. The formation of fibrin at sites of Y. pestis infection supports innate host defense against plague, perhaps by providing a nondiffusible spatial cue that promotes the accumulation of inflammatory cells expressing fibrin-binding integrins. This report demonstrates that fibrin is an essential component of T cell-mediated defense against plague but can be dispensable for Ab-mediated defense. Genetic or pharmacologic depletion of fibrin abrogated innate and T cell-mediated defense in mice challenged intranasally with Y. pestis. The fibrin-deficient mice displayed reduced survival, increased bacterial burden, and exacerbated hemorrhagic pathology. They also showed fewer neutrophils within infected lung tissue and reduced neutrophil viability at sites of liver infection. Depletion of neutrophils from wild-type mice weakened T cell-mediated defense against plague. The data suggest that T cells combat plague in conjunction with neutrophils, which require help from fibrin to withstand Y. pestis encounters and effectively clear bacteria.


Assuntos
Fibrina/fisiologia , Imunidade Inata , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/microbiologia , Yersinia pestis/imunologia , Animais , Proteínas de Bactérias/fisiologia , Fibrinogênio/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peste/imunologia , Peste/metabolismo , Ativadores de Plasminogênio/fisiologia
10.
J Autoimmun ; 42: 71-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23245703

RESUMO

Human herpesviruses establish lifelong latency. Viral recrudescence can lead to the development of cancers, immunoproliferative disorders, transplantation complications, and thrombocytopenia. Although platelet-specific autoantibodies have been reported in patients infected with the Epstein-Barr virus (EBV), the mechanisms by which thrombocytopenia is induced remain unclear, as do the relative contributions of lytic viral replication and latent viral gene expression. The human gammaherpesviruses are tightly restricted in their ability to infect other mammals, so they are difficult to study in live animal models. Here we show that infection of mice with murine gammaherpesvirus-68 (γHV68), a rodent-specific pathogen closely related to EBV, induces the production of platelet-binding antibodies and causes thrombocytopenia. Infection of antibody-deficient mice does not lead to thrombocytopenia, indicating the platelet decrease is mediated by antibody. Additionally, infection with a latency-null recombinant γHV68 does not induce thrombocytopenia, suggesting factors associated with viral latency drive the infection-induced antibody-mediated thrombocytopenia. These studies describe an important animal model of gammaherpesvirus-induced autoimmune thrombocytopenia and demonstrate that this pathology is mediated by antibody and dependent on viral latency. This model will allow studies of the underlying mechanisms of disease progression and the testing of therapeutic strategies for the alleviation of virus-induced thrombocytopenia.


Assuntos
Anticorpos/imunologia , Transtornos Plaquetários/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 4/imunologia , Latência Viral , Animais , Transtornos Plaquetários/etiologia , Plaquetas/imunologia , Células Cultivadas , Modelos Animais de Doenças , Infecções por Vírus Epstein-Barr/complicações , Feminino , Infecções por Herpesviridae/complicações , Humanos , Cadeias mu de Imunoglobulina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade da Espécie , Replicação Viral
11.
J Immunol ; 189(10): 4921-9, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23028058

RESUMO

Influenza causes >250,000 deaths annually in the industrialized world, and bacterial infections frequently cause secondary illnesses during influenza outbreaks, including pneumonia, bronchitis, sinusitis, and otitis media. In this study, we demonstrate that cross-reactive immunity to mismatched influenza strains can reduce susceptibility to secondary bacterial infections, even though this fails to prevent influenza infection. Specifically, infecting mice with H3N2 influenza before challenging with mismatched H1N1 influenza reduces susceptibility to either Gram-positive Streptococcus pneumoniae or Gram-negative Klebsiella pneumoniae. Vaccinating mice with the highly conserved nucleoprotein of influenza also reduces H1N1-induced susceptibility to lethal bacterial infections. Both T cells and Abs contribute to defense against influenza-induced bacterial diseases; influenza cross-reactive T cells reduce viral titers, whereas Abs to nucleoprotein suppress induction of inflammation in the lung. These findings suggest that nonneutralizing influenza vaccines that fail to prevent influenza infection may nevertheless protect the public from secondary bacterial diseases when neutralizing vaccines are not available.


Assuntos
Anticorpos Antivirais/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Infecções por Klebsiella/imunologia , Klebsiella pneumoniae/imunologia , Proteínas do Nucleocapsídeo/imunologia , Infecções por Orthomyxoviridae/imunologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/imunologia , Linfócitos T/imunologia , Animais , Reações Cruzadas , Suscetibilidade a Doenças/imunologia , Suscetibilidade a Doenças/microbiologia , Humanos , Influenza Humana/imunologia , Influenza Humana/microbiologia , Camundongos , Infecções por Orthomyxoviridae/microbiologia
12.
Infect Immun ; 80(1): 206-14, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22064714

RESUMO

Virulence in human-pathogenic Yersinia species is associated with a plasmid-encoded type III secretion system that translocates a set of Yop effector proteins into host cells. One effector, YopE, functions as a Rho GTPase-activating protein (GAP). In addition to acting as a virulence factor, YopE can function as a protective antigen. C57BL/6 mice infected with attenuated Yersinia pestis generate a dominant H2-Kb-restricted CD8 T cell response to an epitope in the N-terminal domain of YopE (YopE69-77), and intranasal vaccination with the YopE69-77 peptide and the mucosal adjuvant cholera toxin (CT) elicits CD8 T cells that are protective against lethal pulmonary challenge with Y. pestis. Because YopE69-77 is conserved in many Yersinia strains, we sought to determine if YopE is a protective antigen for Yersinia pseudotuberculosis and if primary infection with this enteric pathogen elicits a CD8 T cell response to this epitope. Intranasal immunization with the YopE69-77 peptide and CT elicited a CD8 T cell response that was protective against lethal intragastric Y. pseudotuberculosis challenge. The YopE69-77 epitope was a major antigen (∼30% of splenic CD8 T cells were specific for this peptide at the peak of the response) during primary infection with Y. pseudotuberculosis, as shown by flow cytometry tetramer staining. Results of infections with Y. pseudotuberculosis expressing catalytically inactive YopE demonstrated that GAP activity is dispensable for a CD8 T cell response to YopE69-77. Determining the features of YopE that are important for this response will lead to a better understanding of how protective CD8 T cell immunity is generated against Yersinia and other pathogens with type III secretion systems.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Yersinia pseudotuberculosis/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Toxina da Cólera/administração & dosagem , Feminino , Citometria de Fluxo , Epitopos Imunodominantes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia , Análise de Sobrevida , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Infecções por Yersinia pseudotuberculosis/mortalidade , Infecções por Yersinia pseudotuberculosis/prevenção & controle
13.
Infect Immun ; 80(1): 91-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22006565

RESUMO

In mice infected sublethally with Listeria monocytogenes, fibrin is deposited at low levels within hepatic tissue, where it functions protectively by limiting bacterial growth and suppressing hemorrhagic pathology. Here we demonstrate that mice infected with lethal doses of L. monocytogenes produce higher levels of fibrin and display evidence of systemic coagulopathy (i.e., thrombocytopenia, fibrinogen depletion, and elevated levels of thrombin-antithrombin complexes). When the hepatic bacterial burden exceeds 1×10(6) CFU, levels of hepatic fibrin correlate with the bacterial burden, which also correlates with levels of hepatic mRNA encoding the hemostatic enzyme factor XI (FXI). Gene-targeted FXI-deficient mice show significantly improved survival upon challenge with high doses of L. monocytogenes and also display reduced levels of hepatic fibrin, decreased evidence of coagulopathy, and diminished cytokine production (interleukin-6 [IL-6] and IL-10). While fibrin limits the bacterial burden during sublethal listeriosis in wild-type mice, FXI-deficient mice display a significantly improved capacity to restrain the bacterial burden during lethal listeriosis despite their reduced fibrin levels. They also show less evidence of hepatic necrosis. In conjunction with suboptimal antibiotic therapy, FXI-specific monoclonal antibody 14E11 improves survival when administered therapeutically to wild-type mice challenged with high doses of L. monocytogenes. Together, these findings demonstrate the utility of murine listeriosis as a model for dissecting qualitative differences between protective and pathological host responses and reveal novel roles for FXI in exacerbating inflammation and pathogen burden during a lethal bacterial infection.


Assuntos
Deficiência do Fator XI , Listeria monocytogenes/patogenicidade , Listeriose/patologia , Animais , Antibacterianos/uso terapêutico , Anticorpos/uso terapêutico , Coagulação Intravascular Disseminada/microbiologia , Quimioterapia Combinada , Inflamação/patologia , Listeria monocytogenes/crescimento & desenvolvimento , Listeriose/tratamento farmacológico , Listeriose/mortalidade , Fígado/microbiologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sobrevida , Resultado do Tratamento
14.
Vaccine ; 29(44): 7849-56, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-21816194

RESUMO

Influenza A infection induces a massive inflammatory response in the lungs that leads to significant illness and increases the susceptibility to secondary bacterial pneumonia. The most efficient way to prevent influenza infection is through vaccination. While inactivated vaccines induce protective levels of serum antibodies to influenza hemaglutinin (HA) and neuraminidase (NA) surface proteins, these are strain specific and offer little protection against heterosubtypic influenza viruses. In contrast, live attenuated influenza vaccines (LAIVs) induce a T cell response in addition to antibody responses against HA and NA surface proteins. Importantly, LAIV vaccination induces a response in a mouse model that protects against illness due to heterosubtypic influenza strains. While it is not completely clear what is the mechanism of action of LAIV heterosubtypic protection in humans, it has been shown that LAIV induces heterosubtypic protection in mice that is dependent upon a Type 1 immune response and requires CD8 T cells. In this study, we show that LAIV-induced immunity leads to significantly reduced viral titers and inflammatory responses in the lungs of mice following heterosubtypic infection. Not only are viral titers reduced in LAIV vaccinated mice, the amounts of inflammatory cytokines and chemokines in lung tissue are significantly lower. Additionally, we show that LAIV vaccination of healthy adults also induces a robust Type 1 memory response including the production of chemokines and cytokines involved in T cell activation and recruitment. Thus, our results indicate that LAIV vaccination functions by inducing immune memory which can act to modulate the immune response to subsequent heterosubtypic challenge by influencing both innate and adaptive responses.


Assuntos
Proteção Cruzada , Vacinas contra Influenza/imunologia , Adulto , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/análise , Citocinas/imunologia , Experimentação Humana , Humanos , Inflamação/imunologia , Inflamação/patologia , Vacinas contra Influenza/administração & dosagem , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Carga Viral
15.
J Immunol ; 187(4): 1866-76, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21724997

RESUMO

Septic infections dysregulate hemostatic pathways, prompting coagulopathy. Nevertheless, anticoagulant therapies typically fail to protect humans from septic pathology. The data reported in this work may help to explain this discrepancy by demonstrating critical protective roles for coagulation leading to fibrin deposition during host defense against the Gram-negative bacterium Yersinia enterocolitica. After i.p. inoculation with Y. enterocolitica, fibrinogen-deficient mice display impaired cytokine and chemokine production in the peritoneal cavity and suppressed neutrophil recruitment. Moreover, both gene-targeted fibrinogen-deficient mice and wild-type mice treated with the anticoagulant coumadin display increased hepatic bacterial burden and mortality following either i.p. or i.v. inoculation with Y. enterocolitica. Mice with low tissue factor activity succumb to yersiniosis with a phenotype similar to fibrin(ogen)-deficient mice, whereas factor XI-deficient mice show wild-type levels of resistance. Mice deficient in plasminogen activator inhibitor-1 or thrombin-activatable fibrinolysis inhibitor display modest phenotypes, but mice deficient in both plasminogen activator inhibitor-1 and thrombin-activatable fibrinolysis inhibitor succumb to yersiniosis with a phenotype resembling fibrin(ogen)-deficient mice. These findings demonstrate critical protective roles for the tissue factor-dependent extrinsic coagulation pathway during host defense against bacteria and caution that therapeutics targeting major thrombin-generating or antifibrinolytic pathways may disrupt fibrin-mediated host defense during Gram-negative sepsis.


Assuntos
Carboxipeptidase B2/imunologia , Fator XI , Fibrina/imunologia , Serpina E2/imunologia , Tromboplastina/imunologia , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Animais , Carboxipeptidase B2/genética , Carboxipeptidase B2/metabolismo , Fibrina/genética , Fibrina/metabolismo , Humanos , Fígado/imunologia , Fígado/metabolismo , Fígado/microbiologia , Camundongos , Camundongos Knockout , Sepse/genética , Sepse/imunologia , Sepse/metabolismo , Sepse/microbiologia , Sepse/terapia , Serpina E2/genética , Serpina E2/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Yersiniose/genética , Yersiniose/metabolismo , Yersiniose/terapia , Yersinia enterocolitica/metabolismo
16.
J Immunol ; 187(2): 897-904, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21653834

RESUMO

Septic bacterial pneumonias are a major cause of death worldwide. Several of the highest priority bioterror concerns, including anthrax, tularemia, and plague, are caused by bacteria that acutely infect the lung. Bacterial resistance to multiple antibiotics is increasingly common. Although vaccines may be our best defense against antibiotic-resistant bacteria, there has been little progress in the development of safe and effective vaccines for pulmonary bacterial pathogens. The Gram-negative bacterium Yersinia pestis causes pneumonic plague, an acutely lethal septic pneumonia. Historic pandemics of plague caused millions of deaths, and the plague bacilli's potential for weaponization sustains an ongoing quest for effective countermeasures. Subunit vaccines have failed, to date, to fully protect nonhuman primates. In mice, they induce the production of Abs that act in concert with type 1 cytokines to deliver high-level protection; however, the Y. pestis Ags recognized by cytokine-producing T cells have yet to be defined. In this study, we report that Y. pestis YopE is a dominant Ag recognized by CD8 T cells in C57BL/6 mice. After vaccinating with live attenuated Y. pestis and challenging intranasally with virulent plague, nearly 20% of pulmonary CD8 T cells recognize this single, highly conserved Ag. Moreover, immunizing mice with a single peptide, YopE(69-77), suffices to confer significant protection from lethal pulmonary challenge. These findings suggest YopE could be a valuable addition to subunit plague vaccines and provide a new animal model in which sensitive, pathogen-specific assays can be used to study CD8 T cell-mediated defense against acutely lethal bacterial infections of the lung.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Epitopos Imunodominantes/imunologia , Peste/prevenção & controle , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Doença Aguda , Animais , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/patologia , Células Cultivadas , Células Clonais , Modelos Animais de Doenças , Epitopos de Linfócito T/administração & dosagem , Epitopos Imunodominantes/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peste/imunologia , Peste/mortalidade , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/mortalidade , Pneumonia Bacteriana/prevenção & controle , Análise de Sobrevida , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/síntese química , Vacinas Atenuadas/imunologia , Vacinas de Subunidades Antigênicas/síntese química
17.
J Immunol ; 186(3): 1675-84, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21172869

RESUMO

Pneumonic plague is one of the world's most deadly infectious diseases. The causative bacterium, Yersinia pestis, has the potential to be exploited as a biological weapon, and no vaccine is available. Vaccinating B cell-deficient mice with D27-pLpxL, a live attenuated Y. pestis strain, induces cell-mediated protection against lethal pulmonary Y. pestis challenge. In this article, we demonstrate that prime/boost vaccination with D27-pLpxL confers better protection than prime-only vaccination. The improved survival does not result from enhanced bacterial clearance but is associated with increased levels of IL-17 mRNA and protein in the lungs of challenged mice. The boost also increases pulmonary numbers of IL-17-producing CD4 T cells. Interestingly, most of these cells simultaneously produce canonical type 1 and type 17 cytokines; most produce IL-17 and TNF-α, and many produce IL-17, TNF-α, and IFN-γ. Neutralizing IL-17 counteracts the improved survival associated with prime/boost vaccination without significantly impacting bacterial burden. Thus, IL-17 appears to mediate the enhanced protection conferred by booster immunization. Although neutralizing IL-17 significantly reduces neutrophil recruitment to the lungs of mice challenged with Y. pestis, this impact is equally evident in mice that receive one or two immunizations with D27-pLpxL, suggesting it cannot suffice to account for the improved survival that results from booster immunization. We conclude that IL-17 plays a yet to be identified role in host defense that enhances protection against pulmonary Y. pestis challenge, and we suggest that pneumonic plague vaccines should aim to induce mixed type 1 and type 17 cellular responses.


Assuntos
Imunidade Celular , Interleucina-17/fisiologia , Vacina contra a Peste/administração & dosagem , Vacina contra a Peste/imunologia , Peste/imunologia , Peste/prevenção & controle , Yersinia pestis/imunologia , Aciltransferases/administração & dosagem , Aciltransferases/genética , Aciltransferases/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Relação Dose-Resposta Imunológica , Proteínas de Escherichia coli/administração & dosagem , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Imunidade Celular/genética , Esquemas de Imunização , Imunização Secundária/métodos , Interleucina-17/administração & dosagem , Interleucina-17/biossíntese , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Peste/mortalidade , Vacina contra a Peste/genética , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Yersinia pestis/genética
18.
Vaccine ; 29(2): 357-62, 2010 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-20840834

RESUMO

Immunization with the Yersinia pestis F1 and LcrV proteins improves survival in mouse and non-human primate models of pneumonic plague. F1- and LcrV-specific antibodies contribute to protection, however, the mechanisms of antibody-mediated defense are incompletely understood and serum antibody titers do not suffice as quantitative correlates of protection. Previously we demonstrated roles for tumor necrosis factor-alpha (TNFα) and gamma-interferon (IFNγ) during defense against conditionally attenuated pigmentation (pgm) locus-negative Y. pestis. Here, using intranasal challenge with fully virulent pgm-positive Y. pestis strain CO92, we demonstrate that neutralizing TNFα and IFNγ interferes with the capacity of therapeutically administered F1- or LcrV-specific antibody to reduce bacterial burden and increase survival. Moreover, using Y. pestis strain CO92 in an aerosol challenge model, we demonstrate that neutralizing TNFα and IFNγ interferes with protection conferred by immunization with recombinant F1-LcrV fusion protein vaccine (p<0.0005). These findings establish that TNFα and IFNγ contribute to protection mediated by pneumonic plague countermeasures targeting F1 and LcrV, and suggest that an individual's capacity to produce these cytokines in response to Y. pestis challenge will be an important co-determinant of antibody-mediated defense against pneumonic plague.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Interferon gama/imunologia , Vacina contra a Peste/imunologia , Peste/imunologia , Peste/prevenção & controle , Proteínas Citotóxicas Formadoras de Poros/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sobrevida , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
19.
J Exp Med ; 207(3): 591-605, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20212068

RESUMO

RNA splicing is an increasingly recognized regulator of immunity. Here, we demonstrate that after Mycobacterium tuberculosis infection (mRNA) il12rb1 is spliced by dendritic cells (DCs) to form an alternative (mRNA) il12rb1Deltatm that encodes the protein IL-12Rbeta1DeltaTM. Compared with IL-12Rbeta1, IL-12Rbeta1DeltaTM contains an altered C-terminal sequence and lacks a transmembrane domain. Expression of IL-12Rbeta1DeltaTM occurs in CD11c(+) cells in the lungs during M. tuberculosis infection. Selective reconstitution of il12rb1(-/-) DCs with (mRNA) il12rb1 and/or (mRNA) il12rb1Deltatm demonstrates that IL-12Rbeta1DeltaTM augments IL-12Rbeta1-dependent DC migration and activation of M. tuberculosis-specific T cells. It cannot mediate these activities independently of IL12Rbeta1. We hypothesize that M. tuberculosis-exposed DCs express IL-12Rbeta1DeltaTM to enhance IL-12Rbeta1-dependent migration and promote M. tuberculosis-specific T cell activation. IL-12Rbeta1DeltaTM thus represents a novel positive-regulator of IL12Rbeta1-dependent DC function and of the immune response to M. tuberculosis.


Assuntos
Células Dendríticas/imunologia , Mycobacterium tuberculosis/genética , Receptores de Interleucina-12/genética , Processamento Alternativo , Animais , Células da Medula Óssea/fisiologia , Movimento Celular , Quimiocina CCL19/fisiologia , Células Dendríticas/fisiologia , Cinética , Pulmão/imunologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/imunologia , RNA Mensageiro/genética , Receptores de Interleucina-12/deficiência , Receptores de Interleucina-12/imunologia
20.
Infect Immun ; 78(3): 1004-11, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20028811

RESUMO

During inflammatory responses and wound healing, the conversion of soluble fibrinogen to fibrin, an insoluble extracellular matrix, long has been assumed to create a scaffold for the migration of leukocytes and fibroblasts. Previous studies concluded that fibrinogen is a necessary cofactor for mycobacterial trehalose 6,6'-dimycolate-induced responses, because trehalose dimycolate-coated beads, to which fibrinogen was adsorbed, were more inflammatory than those to which other plasma proteins were adsorbed. Herein, we investigate roles for fibrin(ogen) in an in vivo model of mycobacterial granuloma formation and in infection with Mycobacterium tuberculosis, the causative agent of tuberculosis. In wild-type mice, the subcutaneous injection of trehalose dimycolate-coated polystyrene microspheres, suspended within Matrigel, elicited a pyogranulomatous response during the course of 12 days. In fibrinogen-deficient mice, neutrophils were recruited but a more suppurative lesion developed, with the marked degradation and disintegration of the matrix. Compared to that in wild-type mice, the early formation of granulation tissue in fibrinogen-deficient mice was edematous, hypocellular, and disorganized. These deficiencies were complemented by the addition of exogenous fibrinogen. The absence of fibrinogen had no effect on cell recruitment or cytokine production in response to trehalose dimycolate, nor was there a difference in lung histopathology or overall bacterial burden in mice infected with Mycobacterium tuberculosis. In this model, fibrin(ogen) was not required for cell recruitment, cytokine response, or response to infection, but it promoted granulation tissue formation and suppressed leukocyte necrosis.


Assuntos
Fatores Corda/toxicidade , Citocinas/imunologia , Fibrinogênio/imunologia , Leucócitos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Fatores Corda/imunologia , Feminino , Fibrinogênio/genética , Granuloma/patologia , Inflamação/patologia , Pulmão/microbiologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pele/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA