Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
PLoS Pathog ; 14(4): e1006994, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29634758

RESUMO

Zika virus (ZIKV) infection during human pregnancy may cause diverse and serious congenital defects in the developing fetus. Previous efforts to generate animal models of human ZIKV infection and clinical symptoms often involved manipulating mice to impair their Type I interferon (IFN) signaling, thereby allowing enhanced infection and vertical transmission of virus to the embryo. Here, we show that even pregnant mice competent to generate Type I IFN responses that can limit ZIKV infection nonetheless develop profound placental pathology and high frequency of fetal demise. We consistently found that maternal ZIKV exposure led to placental pathology and that ZIKV RNA levels measured in maternal, placental or embryonic tissues were not predictive of the pathological effects seen in the embryos. Placental pathology included trophoblast hyperplasia in the labyrinth, trophoblast giant cell necrosis in the junctional zone, and loss of embryonic vessels. Our findings suggest that, in this context of limited infection, placental pathology rather than embryonic/fetal viral infection may be a stronger contributor to adverse pregnancy outcomes in mice. Our finding demonstrates that in immunocompetent mice, direct viral infection of the embryo is not essential for fetal demise. Our immunologically unmanipulated pregnancy mouse model provides a consistent and easily measurable congenital abnormality readout to assess fetal outcome, and may serve as an additional model to test prophylactic and therapeutic interventions to protect the fetus during pregnancy, and for studying the mechanisms of ZIKV congenital immunopathogenesis.


Assuntos
Modelos Animais de Doenças , Doenças Fetais/patologia , Doenças Placentárias/patologia , Complicações Infecciosas na Gravidez/patologia , Infecção por Zika virus/patologia , Zika virus/fisiologia , Animais , Feminino , Doenças Fetais/virologia , Transmissão Vertical de Doenças Infecciosas , Camundongos , Camundongos Endogâmicos C57BL , Doenças Placentárias/virologia , Gravidez , Complicações Infecciosas na Gravidez/virologia , Resultado da Gravidez , RNA Viral , Infecção por Zika virus/virologia
2.
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
3.
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
4.
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
5.
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
6.
Nat Commun ; 15(1): 200, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172512

RESUMO

The repeat emergence of SARS-CoV-2 variants of concern (VoC) with decreased susceptibility to vaccine-elicited antibodies highlights the need to develop next-generation vaccine candidates that confer broad protection. Here we describe the antibody response induced by the SARS-CoV-2 Spike Ferritin Nanoparticle (SpFN) vaccine candidate adjuvanted with the Army Liposomal Formulation including QS21 (ALFQ) in non-human primates. By isolating and characterizing several monoclonal antibodies directed against the Spike Receptor Binding Domain (RBD), N-Terminal Domain (NTD), or the S2 Domain, we define the molecular recognition of vaccine-elicited cross-reactive monoclonal antibodies (mAbs) elicited by SpFN. We identify six neutralizing antibodies with broad sarbecovirus cross-reactivity that recapitulate serum polyclonal antibody responses. In particular, RBD mAb WRAIR-5001 binds to the conserved cryptic region with high affinity to sarbecovirus clades 1 and 2, including Omicron variants, while mAb WRAIR-5021 offers complete protection from B.1.617.2 (Delta) in a murine challenge study. Our data further highlight the ability of SpFN vaccination to stimulate cross-reactive B cells targeting conserved regions of the Spike with activity against SARS CoV-1 and SARS-CoV-2 variants.


Assuntos
Nanopartículas , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Animais , Camundongos , Anticorpos Neutralizantes , Macaca mulatta , Vacinação , Anticorpos Antivirais , Anticorpos Monoclonais , Vacinas contra COVID-19 , Ferritinas , Glicoproteína da Espícula de Coronavírus/genética
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
J Immunol ; 182(7): 3985-94, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19299696

RESUMO

Natural regulatory T cells (Tregs) constitutively express the IL-2R alpha-chain (CD25) on their surface. Consequently, administration of anti-CD25 Abs is a commonly used technique to deplete Treg populations in vivo. However, activated effector T cells may also transiently express CD25, and are thus also potential targets for anti-CD25 Abs. In this study using Toxoplasma gondii as a model proinflammatory infection, we have examined the capacity of anti-CD25 Abs to target effector T cell populations during an inflammatory episode, to determine to what extent that this action may modulate the outcome of disease. Anti-CD25 Ab-treated C57BL/6 mice displayed significantly reduced CD4(+) T cell IFN-gamma production during acute T. gondii infection and exhibited reduced weight loss and liver pathology during early acute infection; aspects of infection previously associated with effector CD4(+) T cell responses. In agreement, anti-CD25 Ab administration impaired parasite control and caused mice to succumb to infection during late acute/early chronic stages of infection with elevated tissue parasite burdens. In contrast, anti-CD25 Ab treatment of mice with established chronic infections did not markedly affect brain parasite burdens, suggesting that protective T cell populations do not express CD25 during chronic stages of T. gondii infection. In summary, we have demonstrated that anti-CD25 Abs may directly abrogate effector T cell responses during an inflammatory episode, highlighting important limitations of the use of anti-CD25 Ab administration to examine Treg function during inflammatory settings.


Assuntos
Anticorpos Monoclonais/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Toxoplasmose/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Toxoplasmose/patologia
14.
Mucosal Immunol ; 14(3): 703-716, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33214656

RESUMO

Innate lymphoid cells (ILCs) are a heterogeneous family of immune regulators that protect against mucosal pathogens but can also promote intestinal pathology. Although the plasticity between ILCs populations has been described, the role of mucosal pathogens in inducing ILC conversion leading to intestinal pathology remains unclear. Here we demonstrate that IFNγ-producing ILCs are responsible for promoting intestinal pathology in a mouse model of enterocolitis caused by Campylobacter jejuni, a common human enteric pathogen. Phenotypic analysis revealed a distinct population of IFNγ-producing NK1.1-T-bet+ILCs that accumulated in the intestine of C. jejuni-infected mice. Adoptive transfer experiments demonstrated their capacity to promote intestinal pathology. Inactivation of T-bet in NKp46+ ILCs ameliorated disease. Transcriptome analysis and cell-fate mapping experiments revealed that IFNγ-producing NK1.1-ILCs correspond to ILC1 profile and develop from RORγt+ progenitors. Collectively, we identified a distinct population of NK1.1-ex-ILC3s that promotes intestinal pathology through IFNγ production in response to C. jejuni infection.


Assuntos
Infecções por Campylobacter/imunologia , Campylobacter jejuni/fisiologia , Colite/imunologia , Intestinos/imunologia , Linfócitos/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Imunidade Inata , Interferon gama/metabolismo , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Células Th1/imunologia
15.
Infect Immun ; 77(10): 4295-304, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19620344

RESUMO

Vaccinating with live, conditionally attenuated, pigmentation (Pgm)-deficient Yersinia pestis primes T cells that protect mice against pneumonic plague. However, Pgm-deficient strains are not considered safe for human use because they retain substantial virulence in animal models. Y. pestis strains engineered to express Escherichia coli LpxL are avirulent owing to constitutive production of lipopolysaccharide with increased Toll-like receptor 4-activating ability. We generated an LpxL-expressing Pgm-deficient strain (D27-pLpxL) and demonstrate here that this avirulent strain retains the capacity to prime protective T cells. Compared with unvaccinated controls, mice immunized intranasally with live D27-pLpxL exhibit a decreased bacterial burden and increased survival when challenged intranasally with virulent Y. pestis. T cells provide a substantial degree of this protection, as vaccine efficacy is maintained in B-cell-deficient muMT mice unless those animals are depleted of CD4 and CD8 T cells at the time of challenge. Upon challenge with Y. pestis, pulmonary T-cell numbers decline in naive mice, whereas immunized mice show increased numbers of CD44(high) CD43(high) effector T cells and T cells primed to produce tumor necrosis factor alpha and gamma interferon; neutralizing these cytokines at the time of challenge abrogates protection. Immunization does not prevent dissemination of Y. pestis from the lung but limits bacterial growth and pathology in visceral tissue, apparently by facilitating formation of granuloma-like structures. This study describes a new model for studying T-cell-mediated protection against pneumonic plague and demonstrates the capacity for live, highly attenuated, Y. pestis vaccine strains to prime protective memory T-cell responses safely.


Assuntos
Aciltransferases/biossíntese , Vacinas Bacterianas/imunologia , Proteínas de Escherichia coli/biossíntese , Ativação Linfocitária , Peste/prevenção & controle , Linfócitos T/imunologia , Linfócitos T/microbiologia , Yersinia pestis/imunologia , Aciltransferases/genética , Administração Intranasal , Animais , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Contagem de Colônia Microbiana , Proteínas de Escherichia coli/genética , Feminino , Receptores de Hialuronatos/análise , Interferon gama/biossíntese , Leucossialina/análise , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Masculino , Camundongos , Peste/imunologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Análise de Sobrevida , Subpopulações de Linfócitos T/química , Subpopulações de Linfócitos T/imunologia , Linfócitos T/química , Fator de Necrose Tumoral alfa/biossíntese , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Yersinia pestis/genética
17.
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
18.
Cell Host Microbe ; 6(6): 503-12, 2009 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20006839

RESUMO

Surviving infection represents a balance between the proinflammatory responses needed to eliminate the pathogen, and anti-inflammatory signals limiting damage to the host. IL-10 is a potent immunosuppressive cytokine whose impact is determined by the timing and localization of release. We show that NK cells rapidly express IL-10 during acute infection with diverse rapidly disseminating pathogens. The proinflammatory cytokine IL-12 was necessary and sufficient for NK cell induction of IL-10. NK cells from mice with systemic parasitic infection inhibited dendritic cell release of IL-12 in an IL-10-dependent manner, and NK cell depletion resulted in elevated serum IL-12. These data suggest an innate, negative feedback loop in which IL-12 limits its own production by eliciting IL-10 from NK cells. In contrast to disseminating pathogens, locally restricted infections did not elicit NK cell IL-10. Thus systemic infections uniquely engage NK cells in an IL-10-mediated immunoregulatory circuit that functions to alleviate inflammation.


Assuntos
Imunossupressores/imunologia , Infecções/imunologia , Interleucina-10/imunologia , Células Matadoras Naturais/imunologia , Animais , Células Cultivadas , Expressão Gênica , Infecções/microbiologia , Infecções/parasitologia , Interleucina-10/genética , Interleucina-12/genética , Interleucina-12/imunologia , Listeria monocytogenes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Toxoplasma/imunologia , Yersinia pestis/imunologia
19.
Vaccine ; 26(52): 6901-7, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-18926869

RESUMO

Yersinia pestis causes pneumonic plague, an exceptionally virulent disease for which we lack a safe and effective vaccine. Antibodies specific for the Y. pestis F1 and LcrV proteins can protect mice against pulmonary Y. pestis infection. We demonstrate that neutralizing tumor necrosis factor-alpha (TNFalpha) and gamma-interferon (IFNgamma) abrogates this protection at sub-optimal levels of F1- or LcrV-specific antibody, but not at optimal levels. Moreover, we demonstrate that endogenous TNFalpha and IFNgamma confer measurable protection in the complete absence of protective antibodies. These findings indicate that antibodies and cytokines independently protect against pneumonic plague and suggest that surrogate assays for plague vaccine efficacy should consider both the level of vaccine-induced antibody and the capacity of vaccine recipients to produce TNFalpha and IFNgamma upon exposure to Y. pestis.


Assuntos
Anticorpos Antibacterianos/imunologia , Citocinas/uso terapêutico , Vacina contra a Peste/uso terapêutico , Peste/prevenção & controle , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Monoclonais/farmacologia , Antígenos de Bactérias/imunologia , Linfócitos B/fisiologia , Proteínas de Bactérias/imunologia , Relação Dose-Resposta Imunológica , Determinação de Ponto Final , Soros Imunes/farmacologia , Interferon gama/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peste/microbiologia , Vacina contra a Peste/imunologia , Proteínas Citotóxicas Formadoras de Poros/imunologia , Fator de Necrose Tumoral alfa/uso terapêutico , Yersinia pestis/imunologia
20.
Infect Immun ; 75(5): 2630-3, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17307940

RESUMO

Impaired erythropoiesis causes anemia during genetic disorders, chronic disease, and infection. In studies of the underlying mechanisms researchers have increasingly focused on gamma interferon (IFN-gamma). Here, we identified a previously unrecognized role for interleukin-15 (IL-15) in red blood cell homeostasis and demonstrated that IFN-gamma and signal transducer and activator of transcription protein 1-dependent pathways up-regulate expression of IL-15 in vivo. These findings identified new therapeutic targets for anemia.


Assuntos
Anemia , Eritropoese/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-15/metabolismo , Toxoplasma/patogenicidade , Toxoplasmose Animal/fisiopatologia , Anemia/parasitologia , Anemia/fisiopatologia , Animais , Células da Medula Óssea , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição STAT1/metabolismo , Toxoplasmose Animal/parasitologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA