Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
mBio ; 8(6)2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29138299

RESUMO

Although viruses and viral capsids induce rapid immune responses, little is known about viral pathogen-associated molecular patterns (PAMPs) that are exhibited on their surface. Here, we demonstrate that the repeating protein subunit pattern common to most virus capsids is a molecular pattern that induces a Toll-like-receptor-2 (TLR2)-dependent antiviral immune response. This early antiviral immune response regulates the clearance of subsequent bacterial superinfections, which are a primary cause of morbidities associated with influenza virus infections. Utilizing this altered susceptibility to subsequent bacterial challenge as an outcome, we determined that multiple unrelated, empty, and replication-deficient capsids initiated early TLR2-dependent immune responses, similar to intact influenza virus or murine pneumovirus. These TLR2-mediated responses driven by the capsid were not dependent upon the capsid's shape, size, origin, or amino acid sequence. However, they were dependent upon the multisubunit arrangement of the capsid proteins, because unlike intact capsids, individual capsid subunits did not enhance bacterial clearance. Further, we demonstrated that even a linear microfilament protein built from repeating protein subunits (F-actin), but not its monomer (G-actin), induced similar kinetics of subsequent bacterial clearance as did virus capsid. However, although capsids and F-actin induced similar bacterial clearance, in macrophages they required distinct TLR2 heterodimers for this response (TLR2/6 or TLR2/1, respectively) and different phagocyte populations were involved in the execution of these responses in vivo Our results demonstrate that TLR2 responds to invading viral particles that are composed of repeating protein subunits, indicating that this common architecture of virus capsids is a previously unrecognized molecular pattern.IMPORTANCE Rapid and precise pathogen identification is critical for the initiation of pathogen-specific immune responses and pathogen clearance. Bacteria and fungi express common molecular patterns on their exteriors that are recognized by cell surface-expressed host pattern recognition receptors (PRRs) prior to infection. In contrast, viral molecular patterns are primarily nucleic acids, which are recognized after virus internalization. We found that an initial antiviral immune response is induced by the repeating subunit pattern of virus exteriors (capsids), and thus, induction of this response is independent of viral infection. This early response to viral capsids required the cell surface-expressed PRR TLR2 and allowed for improved clearance of subsequent bacterial infection that commonly complicates respiratory viral infections. Since the repeating protein subunit pattern is conserved across viral capsids, this suggests that it is not easy for a virus to change without altering fitness. Targeting this vulnerability could lead to development of a universal antiviral vaccine.


Assuntos
Bactérias/imunologia , Proteínas do Capsídeo/imunologia , Capsídeo/imunologia , Imunidade Inata , Moléculas com Motivos Associados a Patógenos , Receptor 2 Toll-Like/metabolismo , Vírus/imunologia , Animais , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Viroses/imunologia
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
PLoS One ; 4(9): e7142, 2009 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-19774076

RESUMO

BACKGROUND: Destruction of the architectural and subsequently the functional integrity of the lung following pulmonary viral infections is attributable to both the extent of pathogen replication and to the host-generated inflammation associated with the recruitment of immune responses. The presence of antigenically disparate pulmonary viruses and the emergence of novel viruses assures the recurrence of lung damage with infection and resolution of each primary viral infection. Thus, there is a need to develop safe broad spectrum immunoprophylactic strategies capable of enhancing protective immune responses in the lung but which limits immune-mediated lung damage. The immunoprophylactic strategy described here utilizes a protein cage nanoparticle (PCN) to significantly accelerate clearance of diverse respiratory viruses after primary infection and also results in a host immune response that causes less lung damage. METHODOLOGY/PRINCIPAL FINDINGS: Mice pre-treated with PCN, independent of any specific viral antigens, were protected against both sub-lethal and lethal doses of two different influenza viruses, a mouse-adapted SARS-coronavirus, or mouse pneumovirus. Treatment with PCN significantly increased survival and was marked by enhanced viral clearance, accelerated induction of viral-specific antibody production, and significant decreases in morbidity and lung damage. The enhanced protection appears to be dependent upon the prior development of inducible bronchus-associated lymphoid tissue (iBALT) in the lung in response to the PCN treatment and to be mediated through CD4+ T cell and B cell dependent mechanisms. CONCLUSIONS/SIGNIFICANCE: The immunoprophylactic strategy described utilizes an infection-independent induction of naturally occurring iBALT prior to infection by a pulmonary viral pathogen. This strategy non-specifically enhances primary immunity to respiratory viruses and is not restricted by the antigen specificities inherent in typical vaccination strategies. PCN treatment is asymptomatic in its application and importantly, ameliorates the damaging inflammation normally associated with the recruitment of immune responses into the lung.


Assuntos
Brônquios/microbiologia , Brônquios/virologia , Pulmão/virologia , Tecido Linfoide/metabolismo , Nanopartículas/química , Nanotecnologia/métodos , Orthomyxoviridae/metabolismo , Pneumovirus/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Animais , Brônquios/metabolismo , Líquido da Lavagem Broncoalveolar , Coxiella burnetii/metabolismo , Feminino , Infecções/patologia , Pulmão/imunologia , Pulmão/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H
10.
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
11.
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
12.
J Immunol ; 178(2): 1059-67, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17202369

RESUMO

The gut provides a large area for immunization enabling the development of mucosal and systemic Ab responses. To test whether the protective Ags to Yersinia pestis can be orally delivered, the Y. pestis caf1 operon, encoding the F1-Ag and virulence Ag (V-Ag) were cloned into attenuated Salmonella vaccine vectors. F1-Ag expression was controlled under a promoter from the caf1 operon; two different promoters (P), PtetA in pV3, PphoP in pV4, as well as a chimera of the two in pV55 were tested. F1-Ag was amply expressed; the chimera in the pV55 showed the best V-Ag expression. Oral immunization with Salmonella-F1 elicited elevated secretory (S)-IgA and serum IgG titers, and Salmonella-V-Ag(pV55) elicited much greater S-IgA and serum IgG Ab titers than Salmonella-V-Ag(pV3) or Salmonella-V-Ag(pV4). Hence, a new Salmonella vaccine, Salmonella-(F1+V)Ags, made with a single plasmid containing the caf1 operon and the chimeric promoter for V-Ag allowed the simultaneous expression of F1 capsule and V-Ag. Salmonella-(F1+V)Ags elicited elevated Ab titers similar to their monotypic derivatives. For bubonic plague, mice dosed with Salmonella-(F1+V)Ags and Salmonella-F1-Ag showed similar efficacy (>83% survival) against approximately 1000 LD(50) Y. pestis. For pneumonic plague, immunized mice required immunity to both F1- and V-Ags because the mice vaccinated with Salmonella-(F1+V)Ags protected against 100 LD(50) Y. pestis. These results show that a single Salmonella vaccine can deliver both F1- and V-Ags to effect both systemic and mucosal immune protection against Y. pestis.


Assuntos
Antígenos de Bactérias/imunologia , Peste/imunologia , Infecções por Salmonella/imunologia , Infecções por Salmonella/prevenção & controle , Vacinas contra Salmonella/imunologia , Salmonella/imunologia , Yersinia pestis/imunologia , Administração Oral , Animais , Anticorpos/imunologia , Antígenos de Bactérias/genética , Imunogenética , Camundongos , Camundongos Endogâmicos BALB C , Mucosa/imunologia , Peste/prevenção & controle , Salmonella/genética , Vacinas contra Salmonella/administração & dosagem , Taxa de Sobrevida , Yersiniose
13.
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
14.
Chem Biol ; 13(2): 161-70, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16492564

RESUMO

Protein cages, including viral capsids, ferritins, and heat shock proteins (Hsps), can serve as nanocontainers for biomedical applications. They are genetically and chemically malleable platforms, with potential as therapeutic and imaging agent delivery systems. Here, both genetic and chemical strategies were used to impart cell-specific targeting to the Hsp cage from Methanococcus jannaschii. A tumor vasculature targeting peptide was incorporated onto the exterior surface of the Hsp cage. This protein cage bound to alpha(v)beta(3) integrin-expressing cells. Cellular tropism was also imparted by conjugating anti-CD4 antibodies to the exterior of Hsp cages. These Ab-Hsp cage conjugates specifically bound to CD4(+) cells. Protein cages have the potential to simultaneously incorporate multiple functionalities, including cell-specific targeting, imaging, and therapeutic agent delivery. We demonstrate the simultaneous incorporation of two functionalities, imaging and cell-specific targeting, onto the Hsp protein cage.


Assuntos
Proteínas de Choque Térmico/química , Linfócitos/citologia , Melanoma/patologia , Animais , Sequência de Bases , Antígenos CD4/imunologia , Separação Celular , Células Cultivadas , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Mathanococcus/química , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Nanotecnologia
15.
J Immunol ; 174(9): 5462-71, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15843544

RESUMO

Despite the advent of highly active antiretroviral therapy, pulmonary complications in AIDS are a common clinical problem. Pneumocystis jiroveci infection causes a life-threatening pneumonia, especially in individuals with CD4 T cell deficiencies as occurs in AIDS. Although Pneumocystis sp. is an extracellular fungal pathogen, CD8 T cells are the predominant lymphocyte recruited to the lung in CD4-deficient humans and mice during Pneumocystis pneumonia, and we have found that these CD8 T cells are responsible for subsequent lung damage in CD4 T cell-depleted mice. Comparing CD4 T cell-depleted IFN-alpha receptor knockout (KO) mice to wild-type mice, we found that this CD8 T cell recruitment and lung damage is type I IFN (IFN-alphabeta) dependent. However, in both CD4 competent, wild-type and IFN-alpha receptor (IFNAR) KO mice, Pneumocystis infection leads to an eosinophilic granulocyte influx with bronchial epithelial changes as seen in asthma. This response is delayed in IFNAR KO mice, as is pathogen clearance. Although the inflammation is transient in wild-type animals and resolves upon Pneumocystis clearance, it is more severe and persists through day 35 postinfection in IFNAR KO mice, leading to fibrosis. In addition, IFNAR KO, but not wild-type, mice mount a Pneumocystis-specific IgE response, an indicator of allergic sensitization. Thus, in the absence of IFNAR signaling and CD4 T cells, Pneumocystis-mediated lung damage does not occur, whereas in CD4-competent animals, the absence of IFNAR signaling results in an exacerbated Th2 response, asthma-like symptoms, and fibrosis. Therefore, both CD4 T cell- and type I IFN-mediated mechanisms can determine pulmonary complications from Pneumocystis infection.


Assuntos
Interferon-alfa/fisiologia , Interferon beta/fisiologia , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Receptores de Interferon/fisiologia , Transdução de Sinais/imunologia , Alérgenos/imunologia , Animais , Antígenos de Fungos/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Imunoglobulina E/biossíntese , Interferon-alfa/metabolismo , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Pneumonia por Pneumocystis/genética , Pneumonia por Pneumocystis/patologia , Eosinofilia Pulmonar/imunologia , Eosinofilia Pulmonar/microbiologia , Eosinofilia Pulmonar/patologia , Receptor de Interferon alfa e beta , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...