Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bioscience ; 65(2): 183-188, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26834259

RESUMO

Understanding how cultural values influence undergraduate students' science research experiences and career interest is important in efforts to broaden participation and to diversify the biomedical research workforce. The results from our prospective longitudinal study demonstrated that underrepresented minority student (URM) research assistants who see the altruistic value of conducting biomedical research feel more psychologically involved with their research over time, which, in turn, enhances their interest in pursuing a scientific research career. These altruistic motives are uniquely influential to URM students and appear to play an important role in influencing their interest in scientific research careers. Furthermore, seeing how research can potentially affect society and help one's community does not replace typical motives for scientific discovery (e.g., passion, curiosity, achievement), which are important for all students. These findings point to simple strategies for educators, training directors, and faculty mentors to improve retention among undergraduate URM students in biomedicine and the related sciences.

2.
Eur J Immunol ; 44(11): 3263-72, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25091976

RESUMO

Superinfection in mice at day 7 postinfluenza infection exacerbates bacterial pneumonia at least in part via downstream effects of increased IFN-γ signaling. Here we show that up to 3 days postinfluenza infection, mice have reduced susceptibility to superinfection with methicillin-resistant Staphylococcus aureus (MRSA), but that superinfection during that time exacerbated influenza disease. This was due to IL-13 signaling that was advantageous for resolving MRSA infection via inhibition of IFN-γ, but was detrimental to the clearance of influenza virus. However, if superinfection did not occur until the near resolution of influenza infection (day 7), IL-13 signaling was inhibited, at least in part by upregulation of IL-13 decoy receptor (IL-13Rα2), which in turn caused increases in IFN-γ signaling and exacerbation of bacterial infection. Understanding these cytokine sequelae is critical to development of immunotherapies for influenza-MRSA coinfection since perturbations of these sequelae at the wrong time could increase susceptibility to MRSA and/or influenza.


Assuntos
Interferon gama/genética , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Interleucina-13/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções Estafilocócicas/imunologia , Superinfecção/imunologia , Animais , Carga Bacteriana , Coinfecção , Suscetibilidade a Doenças , Interleucina-13/genética , Interleucina-13/farmacologia , Subunidade alfa2 de Receptor de Interleucina-13/antagonistas & inibidores , Subunidade alfa2 de Receptor de Interleucina-13/biossíntese , Pulmão/microbiologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Bacteriana/imunologia , Proteínas Recombinantes/farmacologia , Superinfecção/microbiologia , Carga Viral
3.
Eur J Immunol ; 44(2): 397-408, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24222381

RESUMO

Recent evidence suggests that an individual's unique history and sequence of exposures to pathogens and antigens may dictate downstream immune responses to disparate antigens. We show that the i.n. delivery of nonreplicative virus-like particles (VLPs), which bear structural but no antigenic similarities to respiratory pathogens, acts to prime the lungs of both C56BL/6 and BALB/c mice, facilitating heightened and accelerated primary immune responses to high-dose influenza challenge, thus providing a nonpathogenic model of innate imprinting. These responses correspond closely to those observed following natural infection with the opportunistic fungus, Pneumocystis murina, and are characterized by accelerated antigen processing by DCs and alveolar macrophages, an enhanced influx of cells to the local tracheobronchial lymph node, and early upregulation of T-cell co-stimulatory/adhesion molecules. CD11c⁺ cells, which have been directly exposed to VLPs or Pneumocystis are necessary in facilitating enhanced clearance of influenza virus, and the repopulation of the lung by Ly-6C⁺ precursors relies on CCR2 expression. Thus, immune imprinting 72 h after VLP-priming, or 2 weeks after Pneumocystis-priming is CCR2-mediated and results from the enhanced antigen processing, maturation, and trafficking abilities of DCs and alveolar macrophages, which cause accelerated influenza-specific primary immune responses and result in superior viral clearance.


Assuntos
Antígenos/imunologia , Antígeno CD11c/imunologia , Imunidade Inata/imunologia , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos Ly/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/virologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/virologia , Linfonodos/imunologia , Linfonodos/microbiologia , Linfonodos/virologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Pneumocystis/imunologia , Infecções por Pneumocystis/imunologia , Receptores CCR2/imunologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Linfócitos T/virologia , Regulação para Cima/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia
4.
Lymphat Res Biol ; 11(4): 196-202, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24364842

RESUMO

BACKGROUND: Exposure of the lungs to an antigen or pathogen elicits the formation of lymphoid satellite islands termed inducible bronchus-associated lymphoid tissue (iBALT). However, little is known about how the presence of iBALT, induced by a stimulus unrelated to the subsequent challenge agent, influences systemic immunity in distal locations, whether it be independently, antagonistically, or synergistically. Here, we determined the kinetics of the influenza-specific responses in the iBALT, tracheobronchial lymph node (TBLN), and spleen of mice with and without pre-formed iBALT. METHODS AND RESULTS: Mice with VLP-induced iBALT or no pre-formed iBALT were challenged with influenza. We found that, as we have previously described, those mice whose lungs contained pre-formed iBALT were protected from morbidity, and furthermore, that these mice had increased dendritic cell, and alveolar macrophage accumulation in both the iBALT and TBLNs. This translated to similarly accelerated kinetics and intensified influenza-specific CD4(+), but not CD8(+) T cell responses in the iBALT, TBLN, and spleen. This expansion was then followed by a more rapid T cell contraction in all lymphoid tissues in the mice with pre-formed iBALT. CONCLUSIONS: Thus, iBALT itself may not be responsible for the accelerated primary immune response we observe in mice with pre-formed iBALT, but may contribute to an overall accelerated local and systemic primary CD4(+), but not CD8(+) T cell response. Furthermore, less damaging immune responses observed in mice with pre-formed iBALT may be due to a quicker contraction of CD4(+) T cell responses in both local and systemic secondary lymphoid tissue.


Assuntos
Brônquios/imunologia , Linfócitos T CD4-Positivos/imunologia , Imunidade Celular , Linfonodos/imunologia , Infecções por Orthomyxoviridae/imunologia , Baço/imunologia , Animais , Brônquios/patologia , Brônquios/virologia , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Dendríticas Foliculares/imunologia , Células Dendríticas Foliculares/patologia , Células Dendríticas Foliculares/virologia , Feminino , Imunidade Inata , Vírus da Influenza A Subtipo H1N1/imunologia , Linfonodos/patologia , Linfonodos/virologia , Ativação Linfocitária , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Macrófagos Alveolares/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Baço/patologia , Baço/virologia
5.
ACS Nano ; 7(4): 3036-44, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23540530

RESUMO

Here we present a biomimetic strategy toward nanoparticle design for controlled immune response through encapsulation of conserved internal influenza proteins on the interior of virus-like particles (VLPs) to direct CD8+ cytotoxic T cell protection. Programmed encapsulation and sequestration of the conserved nucleoprotein (NP) from influenza on the interior of a VLP, derived from the bacteriophage P22, results in a vaccine that provides multistrain protection against 100 times lethal doses of influenza in an NP specific CD8+ T cell-dependent manner. VLP assembly and encapsulation of the immunogenic NP cargo protein is the result of a genetically programmed self-assembly making this strategy amendable to the quick production of vaccines to rapidly emerging pathogens. Addition of adjuvants or targeting molecules were not required for eliciting the protective response.


Assuntos
Antígenos de Histocompatibilidade Classe II/administração & dosagem , Nanopartículas/administração & dosagem , Nanopartículas/química , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/síntese química , Antígenos de Histocompatibilidade Classe II/química , Vírus da Influenza A Subtipo H1N1 , Camundongos , Resultado do Tratamento , Vacinas de Partículas Semelhantes a Vírus/química , Vacinas de Partículas Semelhantes a Vírus/imunologia
6.
Am J Pathol ; 181(1): 196-210, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22642909

RESUMO

The importance of the priming of the lung environment by past infections is being increasingly recognized. Exposure to any given antigen can either improve or worsen the outcome of subsequent lung infections, depending on the immunological history of the host. Thus, an ability to impart transient alterations in the lung environment in anticipation of future insult could provide an important novel therapy for emerging infectious diseases. In this study, we show that nasal administration of virus-like particles (VLPs) before, or immediately after, lethal challenge with methicillin-resistant Staphylococcus aureus (MRSA) of mice i) ensures complete recovery from lung infection and near absolute clearance of bacteria within 12 hours of challenge, ii) reduces host response-induced lung tissue damage, iii) promotes recruitment and efficient bacterial clearance by neutrophils and CD11c(+) cells, and iv) protects macrophages from MRSA-induced necrosis. VLP-mediated protection against MRSA relied on innate immunity. Complete recovery occurred in VLP-dosed mice with severe combined immunodeficiency, but not in wild-type mice depleted of either Ly6G(+) or CD11c(+) cells. Early IL-13 production associated with VLP-induced CD11c(+) cells was essential for VLP-induced protection. These results indicate that VLP-induced alteration of the lung environment protects the host from lethal MRSA pneumonia by enhancing phagocyte recruitment and killing and by reducing inflammation-induced tissue damage via IL-13-dependent mechanisms.


Assuntos
Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Fagócitos/imunologia , Pneumonia Estafilocócica/prevenção & controle , Vacinas Antiestafilocócicas/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Imunidade Adaptativa , Administração Intranasal , Animais , Carga Bacteriana , Antígeno CD11c/análise , Células Dendríticas/imunologia , Feminino , Imunofenotipagem , Interleucina-13/imunologia , Pulmão/microbiologia , Macrófagos Alveolares/imunologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Monócitos/imunologia , Neutrófilos/imunologia , Fagocitose/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/patologia , Vacinas Antiestafilocócicas/administração & dosagem , Fatores de Tempo , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem
7.
Vaccine ; 30(24): 3653-65, 2012 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-22465748

RESUMO

We show that a model antigen, ovalbumin (OVA), can be chemically conjugated to the exterior of a small heat shock protein (sHsp) cage that has structural similarities to virus-like particles (VLPs). OVA-sHsp conjugation efficiency was dependent upon the stoichiometry and the length of the small molecule linker utilized, and the attachment position on the sHsp cage. When conjugated OVA-sHsp was delivered intranasally to naïve mice, the resulting immune response to OVA was accelerated and intensified, and OVA-specific IgG1 responses were apparent within 5 days after a single immunizing dose, illustrating its utility for vaccine development. If animals were pretreated with a disparate VLP, P22 (a non-replicative bacteriophage capsid), before OVA-sHsp conjugate immunization, OVA-specific IgG1 responses were apparent already by 4 days after a single immunizing dose of conjugate in OVA-naïve mice. Additionally, the mice pretreated with P22 produced high titer mucosal IgA, and isotype-switched OVA-specific serum IgG. Similarly, sHsp pretreatment enhanced the accumulation of lung germinal center B cells, T follicular helper cells, and increased polymeric Ig receptor expression, priming the lungs for subsequent IgG and IgA responses to influenza virus challenge. Thus, sHsp nanoparticles elicited quick and intense antibody responses and these accelerated responses could similarly be induced to antigen chemically conjugated to the sHsp. Pretreatment of mice with P22 further accelerated the onset of the antibody response to OVA-sHsp, demonstrating the utility of conjugating antigens to VLPs for pre-, or possibly post-exposure prophylaxis of lung, all without the need for adjuvant.


Assuntos
Anticorpos Antivirais/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Nanopartículas , Mucosa Respiratória/imunologia , Vacinação/métodos , Administração Intranasal , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Fatores de Tempo , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/imunologia
8.
Am J Respir Cell Mol Biol ; 46(3): 290-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21960549

RESUMO

It is widely held that exposure to pathogens such as fungi can be an agent of comorbidity, such as exacerbation of asthma or chronic obstructive pulmonary disease. Although many studies have examined allergic responses to fungi and their effects on pulmonary function, the possible pathologic implications of the early innate responses to fungal pathogens have not been explored. We examined early responses to the atypical fungus Pneumocystis in two common strains of mice in terms of overall immunological response and related pathology, such as cell damage and airway hyperresponsiveness (AHR). We found a strong strain-specific response in BALB/c mice that included recruitment of neutrophils, NK, NKT, and CD4 T cells. This response was accompanied by elevated indicators of lung damage (bronchoalveolar lavage fluid albumin and LDH) and profound AHR. This early response was absent in C57BL/6 mice, although both strains exhibited a later response associated with the clearance of Pneumocystis. We found that this AHR could not be attributed exclusively to the presence of recruited neutrophils, NKT, NK, or CD4 cells or to the actions of IFN-γ or IL-4. However, in the absence of STAT6 signaling, AHR and inflammatory cell recruitment were virtually absent. Gene expression analysis indicated that this early response included activation of several transcription factors that could be involved in pulmonary remodeling. These results show that exposure to a fungus such as Pneumocystis can elicit pulmonary responses that may contribute to morbidity, even without prior sensitization, in the context of certain genetic backgrounds.


Assuntos
Hiper-Reatividade Brônquica/metabolismo , Imunidade Inata , Pneumopatias Fúngicas/metabolismo , Pulmão/metabolismo , Infecções por Pneumocystis/metabolismo , Fator de Transcrição STAT6/metabolismo , Albuminas/metabolismo , Animais , Antígenos CD1/genética , Antígenos CD1/metabolismo , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/microbiologia , Hiper-Reatividade Brônquica/fisiopatologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/microbiologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Interferon gama/deficiência , Interferon gama/genética , Interleucina-4/metabolismo , L-Lactato Desidrogenase/metabolismo , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/fisiopatologia , Pneumopatias Fúngicas/genética , Pneumopatias Fúngicas/imunologia , Pneumopatias Fúngicas/microbiologia , Pneumopatias Fúngicas/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/microbiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Infecções por Pneumocystis/genética , Infecções por Pneumocystis/imunologia , Infecções por Pneumocystis/microbiologia , Infecções por Pneumocystis/fisiopatologia , Receptores de Interleucina-4/deficiência , Receptores de Interleucina-4/genética , Receptores de Interleucina-8B/deficiência , Receptores de Interleucina-8B/genética , Fator de Transcrição STAT6/deficiência , Fator de Transcrição STAT6/genética , Transdução de Sinais , Especificidade da Espécie , Fatores de Tempo
9.
PLoS One ; 7(12): e51941, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284825

RESUMO

Coxiella burnetii, the causative agent of Q fever, is a zoonotic disease with potentially life-threatening complications in humans. Inhalation of low doses of Coxiella bacteria can result in infection of the host alveolar macrophage (AM). However, it is not known whether a subset of AMs within the heterogeneous population of macrophages in the infected lung is particularly susceptible to infection. We have found that lower doses of both phase I and phase II Nine Mile C. burnetii multiply and are less readily cleared from the lungs of mice compared to higher infectious doses. We have additionally identified AM resident within the lung prior to and shortly following infection, opposed to newly recruited monocytes entering the lung during infection, as being most susceptible to infection. These resident cells remain infected up to twelve days after the onset of infection, serving as a permissive niche for the maintenance of bacterial infection. A subset of infected resident AMs undergo a distinguishing phenotypic change during the progression of infection exhibiting an increase in surface integrin CD11b expression and continued expression of the surface integrin CD11c. The low rate of phase I and II Nine Mile C. burnetii growth in murine lungs may be a direct result of the limited size of the susceptible resident AM cell population.


Assuntos
Coxiella burnetii/fisiologia , Macrófagos Alveolares/microbiologia , Animais , Carga Bacteriana , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Imunofenotipagem , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Masculino , Camundongos , Fenótipo , Pneumonia/imunologia , Pneumonia/microbiologia , Febre Q/imunologia , Febre Q/microbiologia , Febre Q/mortalidade
10.
Am J Pathol ; 176(6): 2806-18, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395428

RESUMO

Immune-reconstitution after highly active antiretroviral therapy (HAART) is often incomplete, and some HIV-infected individuals fail to regenerate type-I interferon (IFN)-producing pDCs. We recently demonstrated that during Pneumocystis (PC) infection in CD4 T cell-competent mice the absence of type-I IFN signaling results in chronic pulmonary inflammation and fibrosis despite clearance. Because the mechanisms involved are poorly understood, we further characterized the role of type-I IFN signaling in immune responses to PC. We show that type-I IFN signaling around day 7 postinfection is critical to the outcome of inflammation. Microarray analysis of pulmonary CD11c(+) cells revealed that at day 7 post infection, wild-type cells up-regulated type-I IFN-responsive genes as well as SOCS1, which is a critical negative-regulator of type-I IFN and IFN-gamma signaling. This was associated with an eosinophilic lung inflammation, PC clearance, and complete restitution. However, pulmonary CD11c(+) cells from IFNAR(-/-) mice demonstrated increased tumor necrosis factor (TNF)-alpha production and lacked SOCS1-induction at day 7. This was followed by a transient lymphocytic and IFN-gamma response before switching to a chronic eosinophilic inflammation of the lung. Early neutralization of TNF-alpha did not prevent chronic inflammation in IFNAR(-/-) mice, but treatment with an anti-IFN-gamma antibody did. We propose that during PC lung infection type-I IFNs induce SOCS1-associated regulatory mechanisms, which prevent excessive IFN-gamma-mediated responses that cause chronic lung damage. Therefore, partial immune-reconstitution in AIDS, attributable to reduced type-I IFN actions, might disrupt regulatory aspects of inflammation, causing unexplained chronic pulmonary complications as seen in some patients during HAART.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Inflamação , Interferon Tipo I/imunologia , Interferon gama/imunologia , Pulmão , Pneumonia por Pneumocystis , Transdução de Sinais/imunologia , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Antígeno CD11c/genética , Antígeno CD11c/imunologia , Perfilação da Expressão Gênica , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Camundongos SCID , Análise em Microsséries , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/microbiologia , Pneumonia por Pneumocystis/patologia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Fator de Necrose Tumoral alfa/imunologia
11.
Infect Immun ; 78(7): 3019-26, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20351144

RESUMO

The mechanisms of the primary adaptive immune response to Coxiella burnetii are not well known. Following inoculation of the lungs with C. burnetii Nine Mile phase I (NMI), SCID mice developed pneumonia and splenomegaly and succumbed to infection, whereas wild-type mice cleared the infection by 24 days. SCID mice reconstituted with either CD4+ T cells or CD8+ T cells alone were able to control the infection, indicating that the presence of either type of T cells was sufficient to control infection, and B cells were not necessary for primary immunity. Similarly, wild-type mice depleted of either CD4+ T cells or CD8+ T cells controlled infections in their lungs, but these mice were highly susceptible if they were depleted of both types of T cells. However, compared to CD4+ T-cell-dependent protection, CD8+ T-cell-dependent protection resulted in less inflammation in the lungs and less growth of bacteria in the spleens.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Coxiella burnetii/imunologia , Febre Q/imunologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Imunidade Celular/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Febre Q/microbiologia , Febre Q/patologia , Baço/imunologia , Baço/microbiologia , Baço/patologia
12.
Am J Respir Cell Mol Biol ; 42(4): 450-60, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19520922

RESUMO

Influenza virus infections increase susceptibility to secondary bacterial infections, such as pneumococcal pneumonia, resulting in increased morbidity and mortality. Influenza-induced tissue damage is hypothesized to increase susceptibility to Streptococcus pneumoniae infection by increasing adherence to the respiratory epithelium. Using a mouse model of influenza infection followed by S. pneumoniae infection, we found that an influenza infection does not increase the number of pneumococci initially present within the trachea, but does inhibit pneumococcal clearance by 2 hours after infection. To determine whether influenza damage increases pneumococcal adherence, we developed a novel murine tracheal explant system to determine influenza-induced tissue damage and subsequent pneumococcal adherence. Murine tracheas were kept viable ex vivo as shown by microscopic examination of ciliary beating and cellular morphology using continuous media flow for up to 8 days. Tracheas were infected with influenza virus for 0.5-5 days ex vivo, and influenza-induced tissue damage and the early stages of repair to the epithelium were assessed histologically. A prior influenza infection did not increase pneumococcal adherence, even when the basement membrane was maximally denuded or during the repopulation of the basement membrane with undifferentiated epithelial cells. We measured mucociliary clearance in vivo and found it was decreased in influenza-infected mice. Together, our results indicate that exposure of the tracheal basement membrane contributes minimally to pneumococcal adherence. Instead, an influenza infection results in decreased tracheal mucociliary velocity and initial clearance of pneumococci, leading to an increased pneumococcal burden as early as 2 hours after pneumococcal infection.


Assuntos
Vírus da Influenza A Subtipo H1N1/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Infecções Pneumocócicas/metabolismo , Mucosa Respiratória/metabolismo , Streptococcus pneumoniae/metabolismo , Traqueia/metabolismo , Animais , Aderência Bacteriana , Membrana Basal/metabolismo , Membrana Basal/microbiologia , Membrana Basal/patologia , Membrana Basal/virologia , Cílios/metabolismo , Cílios/microbiologia , Cílios/patologia , Cílios/virologia , Feminino , Camundongos , Infecções por Orthomyxoviridae/microbiologia , Infecções por Orthomyxoviridae/patologia , Infecções Pneumocócicas/patologia , Infecções Pneumocócicas/virologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Fatores de Tempo , Traqueia/microbiologia , Traqueia/patologia , Traqueia/virologia
13.
J Infect Dis ; 200(4): 537-45, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19586418

RESUMO

We compared the growth of Streptococcus pneumoniae mutants with a disruption in the gene for either pneumococcal surface protein A (PspA-), neuraminidase A (NanA-), or hyaluronidase (Hyl-) to that of the parental strain D39 by means of a competitive growth model in mice with and those without prior influenza virus infection. The numbers of total bacteria recovered from mice with prior influenza virus infection were significantly greater than those recovered from mice without prior influenza virus infection. Although the Hyl- and NanA- mutants did not display attenuation in mice with or without prior influenza virus infection, the PspA- mutant exhibited attenuation both in mice with and in mice without prior influenza virus infection. This defect was severe in influenza virus-infected mice, for which growth of the PspA- mutant was 1800-fold lower than that of the parental strain D39. Furthermore, PspA immunization significantly reduced secondary bacterial lung burdens and concentrations of specific markers of lung damage in mice receiving serotypes 2, 3, and 4 pneumococci. Our findings indicate that PspA contributes to secondary S. pneumoniae infection after influenza virus infection and that PspA immunization mitigates early secondary pneumococcal lung infections.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Orthomyxoviridae/complicações , Pneumonia Pneumocócica/complicações , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/metabolismo , Animais , Proteínas de Bactérias/genética , Deleção de Genes , Regulação Bacteriana da Expressão Gênica/fisiologia , Hialuronoglucosaminidase/genética , Hialuronoglucosaminidase/metabolismo , Camundongos , Mutação , Neuraminidase/genética , Neuraminidase/metabolismo , Streptococcus pneumoniae/genética
14.
Vaccine ; 27(4): 530-41, 2009 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19026705

RESUMO

Influenza-pseudotyped Gag virus-like particles (VLPs) were produced via the expression of influenza hemagglutinin (HA), neuraminidase (NA) and the murine leukemia virus Gag product in the baculovirus-insect cell expression system. Hemagglutination specific activities of sucrose gradient-purified VLPs were similar to those of egg-grown influenza viruses but particle morphologies were gamma retrovirus-like in the form of consistent 100nm spheres. Immunization of mice and ferrets demonstrated robust immunogenicity and protection from challenge with no measurable morbidity. Ferret data were striking in that immunization with H5N1 VLPs representing either A/Vietnam/1203/04 or A/Indonesia/5/05 resulted in solid protection against highly pathogenic A/Vietnam/1203/04 challenge with no detectable virus in the upper respiratory tract post-challenge in either group. H1N1 VLP immunization of ferrets resulted in partial protection against H5N1 challenge with markedly accelerated virus clearance from the upper respiratory tract relative to controls. The immunogenicity of influenza-pseudotyped VLPs was not dependent on the adjuvant properties of replication competent contaminating baculovirus. These data demonstrate robust vaccine protection of Gag-based, influenza-pseudotyped VLPs carrying a variety of influenza antigens and suggest applicability toward a number of additional respiratory viruses.


Assuntos
Produtos do Gene gag/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vírion/imunologia , Animais , Baculoviridae/imunologia , Células Cultivadas , Feminino , Furões/imunologia , Produtos do Gene gag/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/isolamento & purificação , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuraminidase/imunologia , Infecções por Orthomyxoviridae/imunologia , Fatores de Tempo
15.
J Immunol ; 180(8): 5613-24, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18390746

RESUMO

In contrast to the detrimental outcomes most often associated with the resolution of coinfections, the model presented here involving a localized Pneumocystis infection of the lung, followed 2 wk later by an influenza virus infection, results in a significant beneficial outcome for the host. In the week following the influenza infection, immunocompetent coinfected animals exhibited an accelerated rate of virus clearance, an accelerated appearance of higher influenza-specific neutralizing Ab titers in their serum and bronchoalveolar lavage fluid (BALF), significantly reduced inflammatory cytokine levels in their BALF, and reduced levels of morbidity relative to animals infected only with influenza virus. The beneficial outcome observed in coinfected immunocompetent animals was dependent on the ongoing resolution of a viable Pneumocystis infection. No differences in viral clearance were detected between coinfected and influenza-only-infected muMT mice or likewise for SCID mice. The accelerated anti-influenza response did not appear to be associated with influenza-specific CD8 T cell-mediated responses or NK cell responses in the lung. Rather, the increased rate of viral clearance was due to the enhancement of the influenza-specific Ab response, which in turn was transiently dependent upon the resolution of the ongoing Pneumocystis infection.


Assuntos
Anticorpos Antivirais/imunologia , Citocinas/análise , Vírus da Influenza A Subtipo H1N1/imunologia , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Pneumocystis/imunologia , Albuminas/análise , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/microbiologia , Líquido da Lavagem Broncoalveolar/virologia , Comorbidade , Citocinas/imunologia , Humanos , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , L-Lactato Desidrogenase/análise , Pulmão/microbiologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Pneumocystis/imunologia , Infecções por Pneumocystis/microbiologia
16.
Am J Pathol ; 171(3): 790-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17640969

RESUMO

Improved treatment regimens have reduced fatalities from opportunistic diseases, such as Pneumocystis pneumonia, in AIDS patients. However, serious chronic conditions, including pulmonary hypertension (PH), are increasing in this group. We report here that when CD4 T cells in Pneumocystis-infected mice are temporally depleted and then allowed to return, the extended inflammation results in PH that persists after Pneumocystis is eliminated. Using this model of PH, we have found that i) the onset of PH is correlated with the return of CD4 T cells, but PH persists after CD4 levels diminish; ii) vascular remodeling accompanies PH, but whereas temporary medial hypertrophy is evident with transient PH in immunocompetent mice, persistent PH is associated with perivascular fibrosis; iii) elevated levels of the fibrotic mediator FIZZ1 are found in bronchoalveolar lavage fluid of mice with persistent PH; and iv) although Th2-related mechanisms may be involved in PH etiology, PH still occurs in interleukin-4 receptor-deficient mice under these conditions. Overall, the data presented here demonstrate that the immune response to an infectious disease pathogen, such as Pneumocystis, can, when perturbed and prolonged, lead to later development of a serious chronic condition such as PH.


Assuntos
Hipertensão Pulmonar/etiologia , Pneumonia por Pneumocystis/complicações , Animais , Líquido da Lavagem Broncoalveolar/citologia , Linfócitos T CD4-Positivos/metabolismo , Fibrose/patologia , Infecções por HIV/complicações , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Inflamação/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/patologia , Artéria Pulmonar/citologia , Artéria Pulmonar/patologia , Receptores de Interleucina-4/genética , Receptores de Interleucina-4/metabolismo , Transdução de Sinais/fisiologia
17.
J Parasitol ; 93(3): 562-74, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17626348

RESUMO

Tritrichomonas foetus is the cause of trichomoniasis in cattle. Severe infection is often associated with heavy neutrophil and macrophage accumulation, although it is not known how this response protects during early parasite colonization. The goal of this study was to examine the effects of an early host response upon initial T. foetus colonization within the murine reproductive tract. Mice depleted of neutrophils before T. foetus infection had a significantly higher parasite burden within the reproductive tract compared with mock-depleted control mice. Additionally, gp91(phox-/-)/ iNOS(-/-), and iNOS(-/-) mice had substantially larger parasite burdens than C57BL/6 control mice, whereas gp91l(Phox-/-) mice had similar parasite burden to C57BL/6 control mice. Interestingly, phorbol 12-myristate 13-acetate-stimulated neutrophils and macrophages isolated from all groups of mice were unable to kill T. foetus in vitro. However, macrophages isolated from gp91l(phox-/-) and C57BL/6 mice stimulated with interferon-gamma and lipopolysaccharide were able to kill T. foetus in vitro, whereas macrophages isolated from gp91(phox(-/-)/ iNOS(-/-) and iNOS(-/-) mice were unable to kill T. foetus, suggesting the ability of macrophages to produce reactive nitrogen species but not reactive oxygen species (ROS) is critical for parasite killing during early infection in vivo and in vitro. Additionally, neutrophils seem to control early dissemination of T. foetus throughout the reproductive tract, although production of ROS is not critical for this process.


Assuntos
Neutrófilos/imunologia , Óxido Nítrico Sintase Tipo II/fisiologia , Infecções por Protozoários/imunologia , Espécies Reativas de Nitrogênio/imunologia , Tritrichomonas foetus/imunologia , Animais , Modelos Animais de Doenças , Feminino , Interferon gama/genética , Interferon gama/imunologia , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/imunologia , Útero/parasitologia , Vagina/parasitologia
18.
J Immunol ; 178(10): 6604-15, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17475892

RESUMO

Loss of CD4 T cells is the hallmark of HIV infection. However, type I IFN-producing plasmacytoid dendritic cells may also be lost. This results in susceptibility to an opportunistic infection such as Pneumocystis pneumonia. In addition, regenerative bone marrow failure resulting in pancytopenia is another common problem in advanced stage AIDS. This may be linked to both the failing immune system and recurrent opportunistic infections. We generated lymphocyte-deficient type I IFN receptor-deficient mice (IFrag-/-) to study the effects on Pneumocystis infection of the lung. When IFrag-/- animals were infected with Pneumocystis they died between days 16 and 21 postinfection with minimal pneumonia but severe anemia due to complete bone marrow failure. This included the loss of uncommitted hemopoietic precursor cells. Bone marrow failure was prevented by the reconstitution of IFrag-/- mice with wild-type lymphocytes, especially B cells. T and B cells lacking type I IFN receptor signaling could only partially prevent bone marrow failure in response to Pneumocystis infection. However, the presence of T and B cells lacking type I IFN signaling resulted in compensatory extramedullary hemopoiesis in the liver and spleen. Lymphocyte support of the regenerative capacity of the bone marrow was provided by both type I IFN-dependent and -independent mechanisms that acted synergistically. Our findings point to the requirement of both type I IFNs and lymphocytes in the regenerative capabilities of the hemopoietic system under the pressure of Pneumocystis infection, but not during steady-state hemopoiesis. This may have implications in the management of pancytopenia in AIDS.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Hematopoese Extramedular/imunologia , Interferon-alfa/fisiologia , Interferon beta/fisiologia , Pneumonia por Pneumocystis/imunologia , Pneumonia por Pneumocystis/patologia , Transdução de Sinais/imunologia , Anemia Aplástica/genética , Anemia Aplástica/imunologia , Anemia Aplástica/prevenção & controle , Animais , Subpopulações de Linfócitos B/patologia , Hematopoese Extramedular/genética , Interferon-alfa/deficiência , Interferon-alfa/genética , Interferon beta/deficiência , Interferon beta/genética , Fígado/citologia , Fígado/imunologia , Linfopenia/genética , Linfopenia/imunologia , Linfopenia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Pneumonia por Pneumocystis/sangue , Pneumonia por Pneumocystis/genética , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/genética , Baço/citologia , Baço/imunologia , Baço/transplante
19.
Exp Parasitol ; 115(2): 143-59, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17014850

RESUMO

Environmental stress and endocrine control can affect pathogenesis of sexually transmitted diseases such as trichomoniasis. Acute Tritrichomonas foetus infection was compared in female BALB/c mice to infections in mice treated with high doses of estradiol or housed in constant bright illumination (stressed). In untreated mice, T. foetus readily colonized the reproductive tract, causing minimal epithelial damage and inflammation. Several fold increases of IFN-gamma, TNF-alpha, MCP-1, and IL-6 cytokines were detected after estradiol-treatment of mice, resulting in greatly enhanced inflammation and tissue damage throughout the reproductive tract. Interestingly, estradiol-treatment of mice resulted in reduced T. foetus colonization compared to untreated mice. Infection in stressed mice resulted in increased tissue damage, inflammation, and inflammatory cytokine expression, although parasite colonization within the reproductive tract was similar to that in untreated mice. These results indicate that either estradiol-treatment or stress result in pathogenesis often observed during severe disease. Alternatively, infection in non-treated mice results in chronic colonization, with little inflammation or pathology.


Assuntos
Estradiol/farmacologia , Infecções por Protozoários/etiologia , Estresse Fisiológico/complicações , Tritrichomonas foetus/patogenicidade , Animais , Citocinas/análise , Citocinas/biossíntese , Feminino , Genitália Feminina/imunologia , Genitália Feminina/parasitologia , Genitália Feminina/patologia , Marcação In Situ das Extremidades Cortadas , Luz/efeitos adversos , Linfócitos/efeitos dos fármacos , Linfócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Infecções por Protozoários/imunologia , Infecções por Protozoários/parasitologia , Infecções por Protozoários/patologia , Estresse Fisiológico/etiologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia , Tritrichomonas foetus/efeitos dos fármacos , Tritrichomonas foetus/imunologia
20.
Int J Nanomedicine ; 2(4): 715-33, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18203438

RESUMO

Protein cage nanoparticles have the potential to serve as multifunctional cell targeted, imaging and therapeutic platforms for broad applications in medicine. However, before they find applications in medicine, their biocompatibility in vivo needs to be demonstrated. We provide here baseline biodistribution information of two different spherical protein cage nanoplatforms, the 28 nm viral Cowpea chlorotic mottle virus (CCMV) and the 12 nm heat shock protein (Hsp) cage. In naive and immunized mice both nanoplatforms show similar broad distribution and movement throughout most tissues and organs, rapid excretion, the absence of long-term persistence within mice tissue and organs, and no overt toxicity after a single injection. These results suggest that protein cage based nanoparticles may serve as safe, biocompatible, nanoplatforms for applications in medicine.


Assuntos
Nanopartículas/química , Nanopartículas/ultraestrutura , Proteínas/farmacocinética , Proteínas/ultraestrutura , Animais , Feminino , Taxa de Depuração Metabólica , Camundongos , Camundongos Endogâmicos BALB C , Especificidade de Órgãos , Tamanho da Partícula , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...