Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Infect Immun ; 87(5)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30804102

RESUMEN

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.


Asunto(s)
Fibrinólisis/inmunología , Peste/inmunología , Activadores Plasminogénicos/inmunología , Linfocitos T/inmunología , Factores de Virulencia/inmunología , Yersinia pestis/patogenicidad , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL
2.
PLoS Pathog ; 10(5): e1004142, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24854422

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Celular/genética , Interferón gamma/fisiología , Peste/inmunología , Neumonía Bacteriana/inmunología , Proteínas Citotóxicas Formadoras de Poros/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Yersinia pestis/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peste/complicaciones , Peste/genética , Neumonía Bacteriana/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Factor de Necrosis Tumoral alfa/genética
3.
Clin Infect Dis ; 60(2): 292-7, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25273081

RESUMEN

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.


Asunto(s)
Infecciones por VIH/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Investigación Biomédica/tendencias , Humanos
4.
J Immunol ; 190(8): 4149-61, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23487423

RESUMEN

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.


Asunto(s)
Fibrina/fisiología , Inmunidad Innata , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/microbiología , Yersinia pestis/inmunología , Animales , Proteínas Bacterianas/fisiología , Fibrinógeno/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peste/inmunología , Peste/metabolismo , Activadores Plasminogénicos/fisiología
5.
J Immunol ; 189(10): 4921-9, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23028058

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Proteínas de la Nucleocápside/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Neumocócica/inmunología , Streptococcus pneumoniae/inmunología , Linfocitos T/inmunología , Animales , Reacciones Cruzadas , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/microbiología , Humanos , Gripe Humana/inmunología , Gripe Humana/microbiología , Ratones , Infecciones por Orthomyxoviridae/microbiología
6.
Infect Immun ; 81(6): 2123-32, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23545300

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Oligodesoxirribonucleótidos/administración & dosificación , Peste/prevención & control , Yersinia pestis , Adyuvantes Inmunológicos/farmacología , Administración Intranasal , Animales , Regulación de la Expresión Génica/inmunología , Hígado/microbiología , Pulmón/citología , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/farmacología , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Virulencia , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad
7.
J Autoimmun ; 42: 71-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23245703

RESUMEN

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.


Asunto(s)
Anticuerpos/inmunología , Trastornos de las Plaquetas Sanguíneas/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 4/inmunología , Latencia del Virus , Animales , Trastornos de las Plaquetas Sanguíneas/etiología , Plaquetas/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/complicaciones , Femenino , Infecciones por Herpesviridae/complicaciones , Humanos , Cadenas mu de Inmunoglobulina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de la Especie , Replicación Viral
8.
J Immunol ; 186(3): 1675-84, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21172869

RESUMEN

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.


Asunto(s)
Inmunidad Celular , Interleucina-17/fisiología , Vacuna contra la Peste/administración & dosificación , Vacuna contra la Peste/inmunología , Peste/inmunología , Peste/prevención & control , Yersinia pestis/inmunología , Aciltransferasas/administración & dosificación , Aciltransferasas/genética , Aciltransferasas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Relación Dosis-Respuesta Inmunológica , Proteínas de Escherichia coli/administración & dosificación , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/inmunología , Inmunidad Celular/genética , Esquemas de Inmunización , Inmunización Secundaria/métodos , Interleucina-17/administración & dosificación , Interleucina-17/biosíntesis , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Mutantes , Peste/mortalidad , Vacuna contra la Peste/genética , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Yersinia pestis/genética
9.
J Immunol ; 187(2): 897-904, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21653834

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Epítopos Inmunodominantes/inmunología , Peste/prevención & control , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/patología , Células Cultivadas , Células Clonales , Modelos Animales de Enfermedad , Epítopos de Linfocito T/administración & dosificación , Epítopos Inmunodominantes/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peste/inmunología , Peste/mortalidad , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/mortalidad , Neumonía Bacteriana/prevención & control , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/síntesis química , Vacunas Atenuadas/inmunología , Vacunas de Subunidad/síntesis química
10.
J Immunol ; 187(4): 1866-76, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21724997

RESUMEN

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.


Asunto(s)
Carboxipeptidasa B2/inmunología , Factor XI , Fibrina/inmunología , Serpina E2/inmunología , Tromboplastina/inmunología , Yersiniosis/inmunología , Yersinia enterocolitica/inmunología , Animales , Carboxipeptidasa B2/genética , Carboxipeptidasa B2/metabolismo , Fibrina/genética , Fibrina/metabolismo , Humanos , Hígado/inmunología , Hígado/metabolismo , Hígado/microbiología , Ratones , Ratones Noqueados , Sepsis/genética , Sepsis/inmunología , Sepsis/metabolismo , Sepsis/microbiología , Sepsis/terapia , Serpina E2/genética , Serpina E2/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Yersiniosis/genética , Yersiniosis/metabolismo , Yersiniosis/terapia , Yersinia enterocolitica/metabolismo
11.
Infect Immun ; 80(1): 206-14, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22064714

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Toxina del Cólera/administración & dosificación , Femenino , Citometría de Flujo , Epítopos Inmunodominantes/inmunología , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Análisis de Supervivencia , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Infecciones por Yersinia pseudotuberculosis/mortalidad , Infecciones por Yersinia pseudotuberculosis/prevención & control
12.
Infect Immun ; 80(1): 91-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22006565

RESUMEN

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.


Asunto(s)
Deficiencia del Factor XI , Listeria monocytogenes/patogenicidad , Listeriosis/patología , Animales , Antibacterianos/uso terapéutico , Anticuerpos/uso terapéutico , Coagulación Intravascular Diseminada/microbiología , Quimioterapia Combinada , Inflamación/patología , Listeria monocytogenes/crecimiento & desarrollo , Listeriosis/tratamiento farmacológico , Listeriosis/mortalidad , Hígado/microbiología , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia , Resultado del Tratamiento
13.
Immunol Rev ; 225: 256-71, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18837787

RESUMEN

SUMMARY: Yersinia pestis is one of the world's most virulent human pathogens. Inhalation of this Gram-negative bacterium causes pneumonic plague, a rapidly progressing and usually fatal disease. Extensively antibiotic-resistant strains of Y. pestis exist and have significant potential for exploitation as agents of terrorism and biowarfare. Subunit vaccines comprised of the Y. pestis F1 and LcrV proteins are well-tolerated and immunogenic in humans but cannot be tested for efficacy, because pneumonic plague outbreaks are uncommon and intentional infection of humans is unethical. In animal models, F1/LcrV-based vaccines protect mice and cynomolgus macaques but have failed, thus far, to adequately protect African green monkeys. We lack an explanation for this inconsistent efficacy. We also lack reliable correlate assays for protective immunity. These deficiencies are hampering efforts to improve vaccine efficacy. Here, I review the immunology of pneumonic plague, focusing on evidence that humoral and cellular defense mechanisms collaborate to defend against pulmonary Y. pestis infection.


Asunto(s)
Macrófagos/inmunología , Neutrófilos/inmunología , Vacuna contra la Peste/inmunología , Peste/inmunología , Vacunas de Subunidad/inmunología , Yersinia pestis/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular/inmunología , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/microbiología , Neutrófilos/microbiología , Peste/microbiología , Peste/fisiopatología , Peste/prevención & control , Yersinia pestis/patogenicidad
14.
J Immunol ; 182(7): 3985-94, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19299696

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Toxoplasmosis/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Toxoplasmosis/patología
15.
Infect Immun ; 78(3): 1004-11, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20028811

RESUMEN

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.


Asunto(s)
Factores Cordón/toxicidad , Citocinas/inmunología , Fibrinógeno/inmunología , Leucocitos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Factores Cordón/inmunología , Femenino , Fibrinógeno/genética , Granuloma/patología , Inflamación/patología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piel/patología
16.
J Exp Med ; 197(6): 801-6, 2003 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-12629066

RESUMEN

Fibrin, a product of the blood coagulation cascade, accompanies many type 1 immune responses, including delayed-type hypersensitivity, autoimmunity, and graft rejection. In those settings, fibrin is thought to exacerbate inflammation and disease. Here, we evaluate roles for coagulation during infection with Toxoplasma gondii, a pathogen whose control requires robust type 1 immunity. We establish that fibrin prevents infection-stimulated blood loss, thereby performing a protective function that is essential for survival. Remarkably, fibrin does not simply protect against vascular damage caused directly by the infectious agent, but rather, protects against hemorrhage evoked by interferon-gamma, a critical mediator of type 1 immunity. This finding, to our knowledge, is the first to document a beneficial role for coagulation during type 1 immunity, and suggests that fibrin deposition protects host tissue from collateral damage caused by the immune system as it combats infection.


Asunto(s)
Coagulación Sanguínea/fisiología , Fibrina/metabolismo , Inmunidad/fisiología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Fibrina/genética , Fibrina/inmunología , Interferón gamma/metabolismo , Hígado/metabolismo , Hígado/parasitología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tasa de Supervivencia
17.
J Immunol ; 181(8): 5560-7, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18832714

RESUMEN

At the temperature of its flea vector (approximately 20-30 degrees C), the causative agent of plague, Yersinia pestis, expresses a profile of genes distinct from those expressed in a mammalian host (37 degrees C). When dendritic cells (DC) are exposed to Y. pestis grown at 26 degrees C (Y. pestis-26 degrees), they secrete copious amounts of IL-12p40 homodimer (IL-12(p40)(2)). In contrast, when DCs are exposed to Y. pestis grown at 37 degrees C (Y. pestis-37 degrees), they transcribe very little IL-12p40, which is secreted as IL-12p40 monomer (IL-12p40). Y. pestis-26 degrees also induces migration of DCs to the homeostatic chemokine CCL19, whereas Y. pestis-37 degrees does not; migratory DCs are positive for IL-12p40 transcription and secrete mostly IL-12(p40)(2); DCs lacking IL-12p40 do not migrate. Expression of acyltransferase LpxL from Escherichia coli in Y. pestis-37 degrees results in the production of a hexa-acylated lipid A, also seen in Y. pestis-26 degrees, rather than tetra-acylated lipid A normally seen in Y. pestis-37 degrees. The LpxL-expressing Y. pestis-37 degrees promotes DC IL-12(p40)(2) production and induction of DC migration. In addition, absence of TLR4 ablates production of IL-12(p40)(2) in DC exposed to Y. pestis-26 degrees. The data demonstrate the molecular pathway by which Y. pestis evades induction of early DC activation as measured by migration and IL-12(p40)(2) production.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Regulación Bacteriana de la Expresión Génica/inmunología , Subunidad p40 de la Interleucina-12/inmunología , Peste/inmunología , Receptor Toll-Like 4/inmunología , Yersinia pestis/inmunología , Acetilación , Aciltransferasas/genética , Aciltransferasas/inmunología , Animales , Movimiento Celular/genética , Células Cultivadas , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Células Dendríticas/microbiología , Dimerización , Escherichia coli/genética , Escherichia coli/inmunología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/inmunología , Regulación Bacteriana de la Expresión Génica/genética , Calor , Humanos , Insectos Vectores/microbiología , Subunidad p40 de la Interleucina-12/genética , Lípido A/genética , Lípido A/inmunología , Ratones , Ratones Noqueados , Peste/genética , Siphonaptera/microbiología , Receptor Toll-Like 4/genética , Yersinia pestis/genética
18.
Infect Immun ; 77(10): 4295-304, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19620344

RESUMEN

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.


Asunto(s)
Aciltransferasas/biosíntesis , Vacunas Bacterianas/inmunología , Proteínas de Escherichia coli/biosíntesis , Activación de Linfocitos , Peste/prevención & control , Linfocitos T/inmunología , Linfocitos T/microbiología , Yersinia pestis/inmunología , Aciltransferasas/genética , Administración Intranasal , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Recuento de Colonia Microbiana , Proteínas de Escherichia coli/genética , Femenino , Receptores de Hialuranos/análisis , Interferón gamma/biosíntesis , Leucosialina/análisis , Hígado/inmunología , Hígado/microbiología , Hígado/patología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Peste/inmunología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Análisis de Supervivencia , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/inmunología , Linfocitos T/química , Factor de Necrosis Tumoral alfa/biosíntesis , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Yersinia pestis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA