Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Elife ; 92020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32515732

RESUMO

The squalene-based oil-in-water emulsion (SE) vaccine adjuvant MF59 has been administered to more than 100 million people in more than 30 countries, in both seasonal and pandemic influenza vaccines. Despite its wide use and efficacy, its mechanisms of action remain unclear. In this study we demonstrate that immunization of mice with MF59 or its mimetic AddaVax (AV) plus soluble antigen results in robust antigen-specific antibody and CD8 T cell responses in lymph nodes and non-lymphoid tissues. Immunization triggered rapid RIPK3-kinase dependent necroptosis in the lymph node which peaked at 6 hr, followed by a sequential wave of apoptosis. Immunization with alum plus antigen did not induce RIPK3-dependent signaling. RIPK3-dependent signaling induced by MF59 or AV was essential for cross-presentation of antigen to CD8 T cells by Batf3-dependent CD8+ DCs. Consistent with this, RIPK3 deficient or Batf3 deficient mice were impaired in their ability to mount adjuvant-enhanced CD8 T cell responses. However, CD8 T cell responses were unaffected in mice deficient in MLKL, a downstream mediator of necroptosis. Surprisingly, antibody responses were unaffected in RIPK3-kinase or Batf3 deficient mice. In contrast, antibody responses were impaired by in vivo administration of the pan-caspase inhibitor Z-VAD-FMK, but normal in caspase-1 deficient mice, suggesting a contribution from apoptotic caspases, in the induction of antibody responses. These results demonstrate that squalene emulsion-based vaccine adjuvants induce antigen-specific CD8 T cell and antibody responses, through RIPK3-dependent and-independent pathways, respectively.


Assuntos
Adjuvantes Imunológicos , Formação de Anticorpos , Linfócitos T CD8-Positivos/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Esqualeno/imunologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Emulsões , Imunidade Inata , Linfonodos/citologia , Linfonodos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Polissorbatos , Proteínas Repressoras/metabolismo
2.
Science ; 357(6355): 1014-1021, 2017 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-28798047

RESUMO

Antigen-presenting cells (APCs) occupy diverse anatomical tissues, but their tissue-restricted homeostasis remains poorly understood. Here, working with mouse models of inflammation, we found that mechanistic target of rapamycin (mTOR)-dependent metabolic adaptation was required at discrete locations. mTOR was dispensable for dendritic cell (DC) homeostasis in secondary lymphoid tissues but necessary to regulate cellular metabolism and accumulation of CD103+ DCs and alveolar macrophages in lung. Moreover, while numbers of mTOR-deficient lung CD11b+ DCs were not changed, they were metabolically reprogrammed to skew allergic inflammation from eosinophilic T helper cell 2 (TH2) to neutrophilic TH17 polarity. The mechanism for this change was independent of translational control but dependent on inflammatory DCs, which produced interleukin-23 and increased fatty acid oxidation. mTOR therefore mediates metabolic adaptation of APCs in distinct tissues, influencing the immunological character of allergic inflammation.


Assuntos
Células Dendríticas/imunologia , Homeostase , Hipersensibilidade/metabolismo , Inflamação/metabolismo , Pulmão/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Apresentação de Antígeno , Antígenos CD/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Eosinófilos/imunologia , Ácidos Graxos/metabolismo , Cadeias alfa de Integrinas/metabolismo , Interleucina-23/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Oxirredução , Serina-Treonina Quinases TOR/genética , Células Th17/imunologia , Células Th2/imunologia
3.
Nature ; 531(7595): 523-527, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26982722

RESUMO

The integrated stress response (ISR) is a homeostatic mechanism by which eukaryotic cells sense and respond to stress-inducing signals, such as amino acid starvation. General controlled non-repressed (GCN2) kinase is a key orchestrator of the ISR, and modulates protein synthesis in response to amino acid starvation. Here we demonstrate in mice that GCN2 controls intestinal inflammation by suppressing inflammasome activation. Enhanced activation of ISR was observed in intestinal antigen presenting cells (APCs) and epithelial cells during amino acid starvation, or intestinal inflammation. Genetic deletion of Gcn2 (also known as Eif2ka4) in CD11c(+) APCs or intestinal epithelial cells resulted in enhanced intestinal inflammation and T helper 17 cell (TH17) responses, owing to enhanced inflammasome activation and interleukin (IL)-1ß production. This was caused by reduced autophagy in Gcn2(-/-) intestinal APCs and epithelial cells, leading to increased reactive oxygen species (ROS), a potent activator of inflammasomes. Thus, conditional ablation of Atg5 or Atg7 in intestinal APCs resulted in enhanced ROS and TH17 responses. Furthermore, in vivo blockade of ROS and IL-1ß resulted in inhibition of TH17 responses and reduced inflammation in Gcn2(-/-) mice. Importantly, acute amino acid starvation suppressed intestinal inflammation via a mechanism dependent on GCN2. These results reveal a mechanism that couples amino acid sensing with control of intestinal inflammation via GCN2.


Assuntos
Aminoácidos/metabolismo , Colite/metabolismo , Inflamassomos/antagonistas & inibidores , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Aminoácidos/administração & dosagem , Aminoácidos/deficiência , Aminoácidos/farmacologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Autofagia , Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Colite/etiologia , Colite/patologia , Colite/prevenção & controle , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Humanos , Inflamassomos/metabolismo , Inflamação/etiologia , Inflamação/patologia , Inflamação/prevenção & controle , Interleucina-1beta/imunologia , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico , Células Th17/imunologia , Enzimas Ativadoras de Ubiquitina/deficiência , Enzimas Ativadoras de Ubiquitina/metabolismo
4.
Cell Rep ; 9(5): 1856-1870, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25466255

RESUMO

Matrix metalloproteinase-2 (MMP-2) is involved in several physiological mechanisms, including wound healing and tumor progression. We show that MMP-2 directly stimulates dendritic cells (DCs) to both upregulate OX40L on the cell surface and secrete inflammatory cytokines. The mechanism underlying DC activation includes physical association with Toll-like receptor-2 (TLR2), leading to NF-κB activation, OX40L upregulation on DCs, and ensuing TH2 differentiation. Significantly, MMP-2 polarizes T cells toward type 2 responses in vivo, in a TLR2-dependent manner. MMP-2-dependent type 2 polarization may represent a key immune regulatory mechanism for protection against a broad array of disorders, such as inflammatory, infectious, and autoimmune diseases, which can be hijacked by tumors to evade immunity.


Assuntos
Células Dendríticas/imunologia , Metaloproteinase 2 da Matriz/fisiologia , Receptor 2 Toll-Like/metabolismo , Animais , Células Dendríticas/enzimologia , Células HEK293 , Humanos , Lipopolissacarídeos/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Ligante OX40/metabolismo , Ligação Proteica , Transdução de Sinais , Linfócitos T/enzimologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/fisiologia
5.
Immunity ; 41(3): 478-492, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25220212

RESUMO

Systems biological analysis of immunity to the trivalent inactivated influenza vaccine (TIV) in humans revealed a correlation between early expression of TLR5 and the magnitude of the antibody response. Vaccination of Trl5(-/-) mice resulted in reduced antibody titers and lower frequencies of plasma cells, demonstrating a role for TLR5 in immunity to TIV. This was due to a failure to sense host microbiota. Thus, antibody responses in germ-free or antibiotic-treated mice were impaired, but restored by oral reconstitution with a flagellated, but not aflagellated, strain of E. coli. TLR5-mediated sensing of flagellin promoted plasma cell differentiation directly and by stimulating lymph node macrophages to produce plasma cell growth factors. Finally, TLR5-mediated sensing of the microbiota also impacted antibody responses to the inactivated polio vaccine, but not to adjuvanted vaccines or the live-attenuated yellow fever vaccine. These results reveal an unappreciated role for gut microbiota in promoting immunity to vaccination.


Assuntos
Formação de Anticorpos/imunologia , Vacinas contra Influenza/imunologia , Intestinos/microbiologia , Microbiota/imunologia , Receptor 5 Toll-Like/imunologia , Animais , Anticorpos Antivirais/sangue , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Escherichia coli/imunologia , Flagelina/imunologia , Humanos , Memória Imunológica/imunologia , Influenza Humana/prevenção & controle , Intestinos/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmócitos/imunologia , Plasmócitos/metabolismo , Vacina Antipólio de Vírus Inativado/imunologia , Transdução de Sinais/imunologia , Receptor 5 Toll-Like/biossíntese , Receptor 5 Toll-Like/genética , Vacina contra Febre Amarela/imunologia
6.
BMC Microbiol ; 14: 8, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24428931

RESUMO

BACKGROUND: Visceral leishmaniasis caused by the protozoan parasite Leishmania donovani complex is a potentially fatal disease if left untreated. Few treatment options exist and are toxic, costly and ineffective against resistant strains. Thus a safe and efficacious vaccine to combat this disease is needed. Previously, we reported that intraperitoneal administration of leishmanial antigens (LAg) entrapped in liposomes conferred protection to BALB/c mice against L. donovani challenge infection. However, this vaccine failed to protect mice when administered subcutaneously. We therefore evaluated whether formulation of LAg in combination with two commonly used human-compatible adjuvants, alum and saponin, could improve the protective efficacy of subcutaneously administered LAg, to a level comparable to that of the intraperitoneal liposomal vaccination. RESULTS: Vaccine formulations of LAg with alum or saponin failed to reduce parasite burden in the liver, and alum + LAg immunized mice also failed to reduce parasite burden in the spleen. Interestingly, saponin + LAg vaccination actually resulted in an increased L. donovani parasitic load in the spleen following L. donovani challenge, suggesting this regimen exacerbates the infection. In contrast, mice immunized intraperitoneally with Lip + LAg demonstrated significant protection in both liver and spleen, as expected. Mechanistically, we found that failure of alum + LAg to protect mice was associated with elevated levels of IL-4, whereas both IL-4 and IL-10 levels were increased in saponin + LAg immunized mice. This outcome served to exacerbate L. donovani infection in the saponin + LAg group, despite a concurrent increase in proinflammatory IFN-γ production. On the contrary, protection against L. donovani challenge in Lip + LAg immunized mice was associated with elevated levels of IFN-γ in conjunction with low levels of IL-4 and IL-10 production. CONCLUSIONS: These findings indicate that elevated levels of IL-4 may contribute to LAg vaccine failure, whereas combined elevation of IL-4 together with IL-10 exacerbated the disease as observed in saponin + LAg immunized mice. In contrast, a robust IFN-γ response, in the absence of IL-4 and IL-10 production, was associated with protective immunity following administration of the Lip + LAg vaccine. Together these findings suggest that optimization of antigen/adjuvant formulations to minimize IL-4 and IL-10 induction may be helpful in the development of high efficacy vaccines targeting Leishmania.


Assuntos
Antígenos de Protozoários/imunologia , Interleucina-10/imunologia , Interleucina-4/imunologia , Vacinas contra Leishmaniose/imunologia , Adjuvantes Imunológicos/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Antígenos de Protozoários/administração & dosagem , Modelos Animais de Doenças , Injeções Subcutâneas , Leishmania donovani/isolamento & purificação , Vacinas contra Leishmaniose/administração & dosagem , Leishmaniose Visceral/parasitologia , Leishmaniose Visceral/prevenção & controle , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Carga Parasitária , Saponinas/administração & dosagem , Baço/parasitologia , Falha de Tratamento , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
7.
Science ; 343(6168): 313-317, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24310610

RESUMO

The yellow fever vaccine YF-17D is one of the most successful vaccines ever developed in humans. Despite its efficacy and widespread use in more than 600 million people, the mechanisms by which it stimulates protective immunity remain poorly understood. Recent studies using systems biology approaches in humans have revealed that YF-17D-induced early expression of general control nonderepressible 2 kinase (GCN2) in the blood strongly correlates with the magnitude of the later CD8(+) T cell response. We demonstrate a key role for virus-induced GCN2 activation in programming dendritic cells to initiate autophagy and enhanced antigen presentation to both CD4(+) and CD8(+) T cells. These results reveal an unappreciated link between virus-induced integrated stress response in dendritic cells and the adaptive immune response.


Assuntos
Apresentação de Antígeno , Células Dendríticas/imunologia , Proteínas Serina-Treonina Quinases/biossíntese , Vacina contra Febre Amarela/imunologia , Animais , Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Cricetinae , Células Dendríticas/enzimologia , Ativação Enzimática , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Associadas aos Microtúbulos/genética , Proteínas Serina-Treonina Quinases/genética
8.
Immunol Rev ; 255(1): 243-55, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23947360

RESUMO

For more than a century, immunologists and vaccinologists have existed in parallel universes. Immunologists have for long reveled in using 'model antigens', such as chicken egg ovalbumin or nitrophenyl haptens, to study immune responses in model organisms such as mice. Such studies have yielded many seminal insights about the mechanisms of immune regulation, but their relevance to humans has been questioned. In another universe, vaccinologists have relied on human clinical trials to assess vaccine efficacy, but have done little to take advantage of such trials for studying the nature of immune responses to vaccination. The human model provides a nexus between these two universes, and recent studies have begun to use this model to study the molecular profile of innate and adaptive responses to vaccination. Such 'systems vaccinology' studies are beginning to provide mechanistic insights about innate and adaptive immunity in humans. Here, we present an overview of such studies, with particular examples from studies with the yellow fever and the seasonal influenza vaccines. Vaccination with the yellow fever vaccine causes a systemic acute viral infection and thus provides an attractive model to study innate and adaptive responses to a primary viral challenge. Vaccination with the live attenuated influenza vaccine causes a localized acute viral infection in mucosal tissues and induces a recall response, since most vaccinees have had prior exposure to influenza, and thus provides a unique opportunity to study innate and antigen-specific memory responses in mucosal tissues and in the blood. Vaccination with the inactivated influenza vaccine offers a model to study immune responses to an inactivated immunogen. Studies with these and other vaccines are beginning to reunite the estranged fields of immunology and vaccinology, yielding unexpected insights about mechanisms of viral immunity. Vaccines that have been proven to be of immense benefit in saving lives offer us a new fringe benefit: lessons in viral immunology.


Assuntos
Viroses/imunologia , Viroses/prevenção & controle , Vírus/imunologia , Animais , Humanos , Vacinas Virais/imunologia , Viroses/metabolismo
9.
J Immunol ; 189(6): 2758-67, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22908331

RESUMO

The interaction of CD28, which is constitutively expressed on T cells, with B7.1/B7.2 expressed on APCs is critical for T cell activation. CD28 is also expressed on murine and human plasma cells but its function on these cells remains unclear. There are two types of plasma cells: short-lived ones that appear in the secondary lymphoid tissue shortly after Ag exposure, and long-lived plasma cells that mainly reside in the bone marrow. We demonstrate that CD28-deficient murine short- and long-lived plasma cells produce significantly higher levels of Abs than do their wild-type counterparts. This was owing to both increased frequencies of plasma cells as well as increased Ab production per plasma cell. Plasma cells also express the ligand for CD28, B7.1, and B7.2. Surprisingly, deficiency of B7.1 and B7.2 in B cells also led to higher Ab levels, analogous to Cd28(-/-) plasma cells. Collectively, our results suggest that the CD28-B7 interaction operates as a key modulator of plasma cell function.


Assuntos
Antígenos B7/fisiologia , Antígenos CD28/fisiologia , Sobrevivência Celular/imunologia , Senescência Celular/imunologia , Plasmócitos/citologia , Plasmócitos/imunologia , Animais , Antígenos B7/deficiência , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Antígenos CD28/biossíntese , Antígenos CD28/deficiência , Linhagem Celular , Feminino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Plasmócitos/metabolismo , Baço/citologia , Baço/imunologia , Baço/metabolismo , Fatores de Tempo
10.
Mol Pharm ; 9(1): 59-70, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22133194

RESUMO

The development of a long-term protective subunit vaccine against visceral leishmaniasis depends on antigens and adjuvants that can induce an appropriate immune response. The immunization of leishmanial antigens alone shows limited efficacy in the absence of an appropriate adjuvant. Earlier we demonstrated sustained protection against Leishmania donovani with leishmanial antigens entrapped in cationic liposomes through an intraperitoneal route. However, this route is not applicable for human administration. Herein, we therefore evaluated the immune response and protection induced by liposomal soluble leishmanial antigen (SLA) formulated with monophosphoryl lipid-trehalose dicorynomycolate (MPL-TDM) through a subcutaneous route. Subcutaneous immunization of BALB/c mice with SLA entrapped in liposomes or with MPL-TDM elicited partial protection against experimental visceral leishmaniasis. In contrast, liposomal SLA adjuvanted with MPL-TDM induced significantly higher levels of protection in liver and spleen in BALB/c mice challenged 10 days post-vaccination. Protection conferred by this formulation was sustained up to 12 weeks of immunization, and infection was controlled for at least 4 months of the challenge, similar to liposomal SLA immunization administered intraperitoneally. An analysis of cellular immune responses of liposomal SLA + MPL-TDM immunized mice demonstrated the induction of IFN-γ and IgG2a antibody production not only 10 days or 12 weeks post-vaccination but also 4 months after the challenge infection and a down regulation of IL-4 production after infection. Moreover, long-term immunity elicited by this formulation was associated with IFN-γ production also by CD8⁺ T cells. Taken together, our results suggest that liposomal SLA + MPL-TDM represent a good vaccine formulation for the induction of durable protection against L. donovani through a human administrable route.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Protozoários/administração & dosagem , Fatores Corda/administração & dosagem , Leishmania donovani/imunologia , Vacinas contra Leishmaniose/administração & dosagem , Leishmaniose Visceral/prevenção & controle , Lipídeo A/análogos & derivados , Fosfolipídeos/administração & dosagem , Adjuvantes Imunológicos/química , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/isolamento & purificação , Antígenos de Protozoários/uso terapêutico , Células Cultivadas , Fenômenos Químicos , Fatores Corda/química , Imunidade Ativa , Imunidade Celular , Memória Imunológica , Injeções Subcutâneas , Interferon gama/metabolismo , Interleucina-4/metabolismo , Leishmania donovani/metabolismo , Vacinas contra Leishmaniose/química , Vacinas contra Leishmaniose/imunologia , Vacinas contra Leishmaniose/uso terapêutico , Leishmaniose Visceral/imunologia , Leishmaniose Visceral/parasitologia , Lipídeo A/administração & dosagem , Lipídeo A/química , Lipossomos , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Fosfolipídeos/química , Organismos Livres de Patógenos Específicos , Baço/imunologia , Baço/metabolismo , Baço/parasitologia , Baço/patologia
11.
Nature ; 470(7335): 543-7, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21350488

RESUMO

Many successful vaccines induce persistent antibody responses that can last a lifetime. The mechanisms by which they do so remain unclear, but emerging evidence indicates that they activate dendritic cells via Toll-like receptors (TLRs). For example, the yellow fever vaccine YF-17D, one of the most successful empiric vaccines ever developed, activates dendritic cells via multiple TLRs to stimulate proinflammatory cytokines. Triggering specific combinations of TLRs in dendritic cells can induce synergistic production of cytokines, which results in enhanced T-cell responses, but its impact on antibody responses remain unknown. Learning the critical parameters of innate immunity that program such antibody responses remains a major challenge in vaccinology. Here we demonstrate that immunization of mice with synthetic nanoparticles containing antigens plus ligands that signal through TLR4 and TLR7 induces synergistic increases in antigen-specific, neutralizing antibodies compared to immunization with nanoparticles containing antigens plus a single TLR ligand. Consistent with this there was enhanced persistence of germinal centres and of plasma-cell responses, which persisted in the lymph nodes for >1.5 years. Surprisingly, there was no enhancement of the early short-lived plasma-cell response relative to that observed with single TLR ligands. Molecular profiling of activated B cells, isolated 7 days after immunization, indicated that there was early programming towards B-cell memory. Antibody responses were dependent on direct triggering of both TLRs on B cells and dendritic cells, as well as on T-cell help. Immunization protected completely against lethal avian and swine influenza virus strains in mice, and induced robust immunity against pandemic H1N1 influenza in rhesus macaques.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Imunidade Inata/imunologia , Memória Imunológica/imunologia , Vacinas contra Influenza/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Células Dendríticas/citologia , Células Dendríticas/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/administração & dosagem , Ácido Láctico , Ligantes , Linfonodos/citologia , Linfonodos/imunologia , Ativação Linfocitária , Macaca mulatta/imunologia , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Plasmócitos/citologia , Plasmócitos/imunologia , Plasmócitos/metabolismo , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Linfócitos T/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
12.
Science ; 329(5993): 849-53, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20705860

RESUMO

Dendritic cells (DCs) play a vital role in initiating robust immunity against pathogens as well as maintaining immunological tolerance to self antigens. However, the intracellular signaling networks that program DCs to become tolerogenic remain unknown. We report here that the Wnt-beta-catenin signaling in intestinal dendritic cells regulates the balance between inflammatory versus regulatory responses in the gut. beta-catenin in intestinal dendritic cells was required for the expression of anti-inflammatory mediators such as retinoic acid-metabolizing enzymes, interleukin-10, and transforming growth factor-beta, and the stimulation of regulatory T cell induction while suppressing inflammatory effector T cells. Furthermore, ablation of beta-catenin expression in DCs enhanced inflammatory responses and disease in a mouse model of inflammatory bowel disease. Thus, beta-catenin signaling programs DCs to a tolerogenic state, limiting the inflammatory response.


Assuntos
Células Dendríticas/imunologia , Inflamação , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Tolerância a Antígenos Próprios , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , beta Catenina/metabolismo , Animais , Citocinas/metabolismo , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/citologia , Tretinoína/metabolismo , Proteínas Wnt/metabolismo
13.
BMC Microbiol ; 10: 181, 2010 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-20576102

RESUMO

BACKGROUND: The development of an effective vaccine against visceral leishmaniasis (VL) caused by Leishmania donovani is an essential aim for controlling the disease. Use of the right adjuvant is of fundamental importance in vaccine formulations for generation of effective cell-mediated immune response. Earlier we reported the protective efficacy of cationic liposome-associated L. donovani promastigote antigens (LAg) against experimental VL. The aim of the present study was to compare the effectiveness of two very promising adjuvants, Bacille Calmette-Guerin (BCG) and Monophosphoryl lipid A (MPL) plus trehalose dicorynomycolate (TDM) with cationic liposomes, in combination with LAg, to confer protection against murine VL. RESULTS: All the three formulations afforded significant protection against L. donovani in both the visceral organs, liver and spleen. Although comparable level of protection was observed in BCG+LAg and MPL-TDM+LAg immunized mice, highest level of protection was exhibited by the liposomal LAg immunized group. Significant increase in anti-LAg IgG levels were detected in both MPL-TDM+LAg and liposomal LAg immunized animals with higher levels of IgG2a than IgG1. But BCG+LAg failed to induce any antibody response. As an index of cell-mediated immunity DTH responses were measured and significant response was observed in mice vaccinated with all the three different formulations. However, highest responses were observed with liposomal vaccine immunization. Comparative evaluation of IFN-gamma and IL-4 responses in immunized mice revealed that MPL-TDM+LAg group produced the highest level of IFN-gamma but lowest IL-4 level, while BCG+LAg demonstrated generation of suboptimum levels of both IFN-gamma and IL-4 response. Elicitation of moderate levels of prechallenge IFN-gamma along with optimum IL-4 corresponds with successful vaccination with liposomal LAg. CONCLUSION: This comparative study reveals greater effectiveness of the liposomal vaccine for protection against progressive VL in BALB/c. Again, evaluation of the immune responses by vaccination emphasizes the need of stimulation of potent cellular immunity based on both Th1 and Th2 cell responses to confer protection against VL.


Assuntos
Antígenos de Protozoários/imunologia , Vacina BCG/imunologia , Vacinas contra Leishmaniose/imunologia , Leishmaniose Visceral/prevenção & controle , Lipídeo A/análogos & derivados , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antiprotozoários/sangue , Fatores Corda/farmacologia , Feminino , Imunidade Humoral , Lipídeo A/farmacologia , Camundongos , Fatores de Tempo
14.
Nat Immunol ; 11(7): 608-17, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20495560

RESUMO

The mechanisms that initiate T helper type 2 (T(H)2) responses are poorly understood. Here we demonstrate that cysteine protease-induced T(H)2 responses occur via 'cooperation' between migratory dermal dendritic cells (DCs) and basophils positive for interleukin 4 (IL-4). Subcutaneous immunization with papain plus antigen induced reactive oxygen species (ROS) in lymph node DCs and in dermal DCs and epithelial cells of the skin. ROS orchestrated T(H)2 responses by inducing oxidized lipids that triggered the induction of thymic stromal lymphopoietin (TSLP) by epithelial cells mediated by Toll-like receptor 4 (TLR4) and the adaptor protein TRIF; by suppressing production of the T(H)1-inducing molecules IL-12 and CD70 in lymph node DCs; and by inducing the DC-derived chemokine CCL7, which mediated recruitment of IL-4(+) basophils to the lymph node. Thus, the T(H)2 response to cysteine proteases requires DC-basophil cooperation via ROS-mediated signaling.


Assuntos
Basófilos/metabolismo , Citocinas/biossíntese , Células de Langerhans/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células Th2/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antígenos/imunologia , Basófilos/imunologia , Basófilos/patologia , Comunicação Celular , Citocinas/genética , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Interleucina-4/biossíntese , Células de Langerhans/imunologia , Células de Langerhans/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Papaína/imunologia , Espécies Reativas de Oxigênio/imunologia , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Linfopoietina do Estroma do Timo
15.
Nat Med ; 15(4): 401-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19252500

RESUMO

Immune sensing of a microbe occurs via multiple receptors. How signals from different receptors are coordinated to yield a specific immune response is poorly understood. We show that two pathogen recognition receptors, Toll-like receptor 2 (TLR2) and dectin-1, recognizing the same microbial stimulus, stimulate distinct innate and adaptive responses. TLR2 signaling induced splenic dendritic cells (DCs) to express the retinoic acid metabolizing enzyme retinaldehyde dehydrogenase type 2 and interleukin-10 (IL-10) and to metabolize vitamin A and stimulate Foxp3(+) T regulatory cells (T(reg) cells). Retinoic acid acted on DCs to induce suppressor of cytokine signaling-3 expression, which suppressed activation of p38 mitogen-activated protein kinase and proinflammatory cytokines. Consistent with this finding, TLR2 signaling induced T(reg) cells and suppressed IL-23 and T helper type 17 (T(H)17) and T(H)1-mediated autoimmune responses in vivo. In contrast, dectin-1 signaling mostly induced IL-23 and proinflammatory cytokines and augmented T(H)17 and T(H)1-mediated autoimmune responses in vivo. These data define a new mechanism for the systemic induction of retinoic acid and immune suppression against autoimmunity.


Assuntos
Autoimunidade/imunologia , Células Dendríticas/imunologia , Proteínas de Membrana/imunologia , Proteínas do Tecido Nervoso/imunologia , Linfócitos T Reguladores/imunologia , Receptor 2 Toll-Like/imunologia , Vitamina A/metabolismo , Aldeído Desidrogenase/metabolismo , Animais , Interleucina-10/imunologia , Interleucina-23/imunologia , Lectinas Tipo C , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais , Baço/imunologia , Linfócitos T Auxiliares-Indutores/imunologia
16.
Infect Immun ; 76(3): 1003-15, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18195029

RESUMO

Visceral leishmaniasis is deadly if not treated, and development of a vaccine with long-term immunity remains a challenge. In this study, we showed that cationic distearoyl phosphatidylcholine (DSPC) liposomes, when used as vaccine adjuvant with the immunodominant 63-kDa glycoprotein (gp63) of Leishmania donovani promastigotes, induced significant protection against progressive visceral leishmaniasis in susceptible BALB/c mice. gp63 used without adjuvant elicited partial protection but in association with liposomes exhibited marked resistance in both the livers and spleens of the mice challenged 10 days after the last vaccination. The protective efficacy of liposomal gp63 vaccination was dose dependent, with 2.5 mug of protein showing optimal protection. The immunity conferred by this vaccine formulation was durable, as mice challenged 12 weeks after immunization were still protected, and the infection was controlled for at least 3 months postchallenge. Production of gamma interferon (IFN-gamma) and interleukin-4 (IL-4) by splenic T cells, and of serum immunoglobulin G1 (IgG1) and IgG2a following immunization, suggested that a mixed Th1/Th2 response had been induced following immunization. However, control of disease progression and parasitic burden in mice vaccinated with gp63 in cationic DSPC liposomes was associated with enhancement of antigen-specific IFN-gamma and downregulation of IL-4, demonstrating a Th1 bias. Long-term immunity elicited by this vaccine corresponded to, in addition to the presence of antigen-specific Th1, CD8+ T-cell responses. Our results demonstrated that stable cationic liposomes containing gp63 acted as a potent adjuvant for protein antigen to induce long-term protection against L. donovani that represents an alternative to DNA vaccination.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Protozoários/imunologia , Vacinas contra Leishmaniose/imunologia , Leishmaniose Visceral/prevenção & controle , Lipossomos/administração & dosagem , Metaloendopeptidases/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antiprotozoários/sangue , Linfócitos T CD8-Positivos/imunologia , Relação Dose-Resposta Imunológica , Imunoglobulina G/sangue , Interferon gama/metabolismo , Interleucina-4/metabolismo , Leishmania donovani/imunologia , Leishmaniose Visceral/imunologia , Lipossomos/farmacologia , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Fosfatidilcolinas/administração & dosagem , Fosfatidilcolinas/farmacologia , Baço/parasitologia , Subpopulações de Linfócitos T/imunologia , Células Th1/imunologia , Fatores de Tempo
17.
Vaccine ; 25(52): 8771-81, 2007 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-18031874

RESUMO

The difficulty in making successful vaccines against leishmaniasis is partly due to lack of an appropriate adjuvant. Non-coding plasmid DNA (pDNA) bearing immunostimulatory sequences (ISS) is a potent activator of innate immunity, and can thus act as an adjuvant with vaccine antigen. We therefore evaluated the efficacy of pDNA and soluble leishmanial antigens (SLA) to protect against challenge with Leishmania donovani infection. We demonstrate that immunomodulatory activity of pDNA, which potentiated a Th1 immune responses, led to enhanced protection with SLA. Importantly, adding cationic liposomes as vehicle to the antigen, with pDNA either complexed or entrapped within, significantly increased the potentiating effect of pDNA. Further, comparison of the two vaccine formulations demonstrated an impressive increase in the protective efficacy up to two folds when both antigen and pDNA were within the vehicle. Thus, these studies establish the utility of non-coding pDNA bearing ISS as strong promoters of vaccine potency of liposomal antigens especially when co-entrapped with the antigen in cationic liposomes.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Protozoários/imunologia , DNA Bacteriano/imunologia , Leishmania donovani/imunologia , Leishmaniose Visceral/prevenção & controle , Lipossomos/imunologia , Vacinas Protozoárias/imunologia , Animais , Citocinas/biossíntese , Feminino , Leishmaniose Visceral/imunologia , Lipossomos/administração & dosagem , Fígado/parasitologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/imunologia , Baço/parasitologia , Células Th1/imunologia
18.
J Immunol ; 179(9): 6169-75, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17947692

RESUMO

Bacterial flagellin is a target of innate and adaptive immune responses during Salmonella infection. Intravenous injection of Salmonella flagellin into C57BL/6 mice induced rapid IL-6 production and increased expression of activation markers by splenic dendritic cells. CD11b(+), CD8alpha(+), and plasmacytoid dendritic cells each increased expression of CD86 and CD40 in response to flagellin stimulation, although CD11b(+) dendritic cells were more sensitive than the other subsets. In addition, flagellin caused the rapid redistribution of dendritic cells from the red pulp and marginal zone of the spleen into the T cell area of the white pulp. Purified splenic dendritic cells did not respond directly to flagellin, indicating that flagellin-mediated activation of splenic dendritic cells occurs via bystander activation. IL-6 production, increased expression of activation markers, and dendritic cell redistribution in the spleen were dependent on MyD88 expression by bone marrow-derived cells. Avoiding this innate immune response to flagellin is important for bacterial survival, because Salmonella-overexpressing recombinant flagellin was highly attenuated in vivo. These data indicate that flagellin-mediated activation of dendritic cells is rapid, mediated by bystander activation, and highly deleterious to bacterial survival.


Assuntos
Efeito Espectador/imunologia , Replicação do DNA/genética , DNA Bacteriano/genética , Células Dendríticas/imunologia , Flagelina/imunologia , Salmonella/imunologia , Baço/imunologia , Animais , Antígeno CD11b/metabolismo , Antígenos CD8/metabolismo , Linhagem Celular , Interleucina-6/sangue , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Salmonella/imunologia
19.
J Immunol ; 179(8): 5592-603, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17911647

RESUMO

Visceral leishmaniasis (VL) or kala-azar is known to be associated with a mixed Th1-Th2 response, and effective host defense requires the induction of IFN-gamma and IL-12. We address the role of the differential decline of IL-10 and TGF-beta in response to sodium antimony gluconate (SAG) and amphotericin B (AmB), the therapeutic success of SAG and AmB in Indian VL, and the significance of IL-10 and TGF-beta in the development and progression of post-kazla-azar dermal leishmaniasis (PKDL). In the active disease, PBMC from VL patients showed suppressed Ag-specific lymphoproliferation, IFN-gamma and IL-12 production, and elevation of IL-10 and TGF-beta. Cure corresponded with an elevation in IFN-gamma and IL-12 production and down-regulation of IL-10 and TGF-beta. Both CD4(+) and CD8(+) T cells were involved in IFN-gamma and IL-10 production. Interestingly, the retention and maintenance of residual IL-10 and TGF-beta in some SAG-treated individuals and the elevation of IL-10 and TGF-beta in PKDL, a sequel to kala-azar, probably reflects the role of these cytokines in reactivation of the disease in the form of PKDL. Contrastingly, AmB treatment of VL resulted in negligible TGF-beta levels and absolute elimination of IL-10, reflecting the better therapeutic activity of AmB and its probable role in the recent decline in PKDL occurrences in India. Moreover, elucidation of immune responses in Indian PKDL patients revealed a spectral pattern of disease progression where disease severity could be correlated inversely with lymphoproliferation and directly with TGF-beta, IL-10, and Ab production. In addition, the enhancement of CD4(+)CD25(+) T cells in active VL, their decline at cure, and reactivation in PKDL suggest their probable immunosuppressive role in these disease forms.


Assuntos
Anfotericina B/uso terapêutico , Suscetibilidade a Doenças/imunologia , Interleucina-10/fisiologia , Leishmania donovani/imunologia , Leishmaniose Cutânea/tratamento farmacológico , Leishmaniose Cutânea/imunologia , Leishmaniose Visceral/imunologia , Fator de Crescimento Transformador beta/fisiologia , Adolescente , Adulto , Animais , Gluconato de Antimônio e Sódio/uso terapêutico , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Índia/epidemiologia , Leishmania donovani/efeitos dos fármacos , Leishmaniose Cutânea/epidemiologia , Leishmaniose Visceral/tratamento farmacológico , Leishmaniose Visceral/epidemiologia , Masculino , Recidiva
20.
Proc Natl Acad Sci U S A ; 104(29): 12075-80, 2007 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-17615242

RESUMO

The contribution of CCR6 and phagocyte recruitment to the initiation of T cell responses to a local pathogen is unclear. CD4 T cell activation to an injected soluble antigen occurred rapidly and was completely CCR6-independent. In marked contrast, the tempo of pathogen-specific CD4 T cell activation depended on whether the antigen was secreted or cell-associated. Furthermore, lymph node pathogen-specific CD4 T cell activation required CCR6 and cell migration from the site of infection. Surprisingly, adoptive transfer of wild-type blood phagocytes rescued bacteria-specific T cell activation in CCR6-deficient mice, even when these cells were unable to participate in direct antigen presentation. These data demonstrate that T cell responses to a local bacterial infection follow a distinct tempo, largely determined by bacterial protein secretion, and that CCR6-mediated blood phagocyte recruitment to the site of infection is a critical step in the initiation of pathogen-specific immune responses in skin draining lymph nodes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Movimento Celular , Fagócitos/citologia , Fagócitos/imunologia , Receptores de Quimiocinas/imunologia , Infecções por Salmonella/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/imunologia , Antígenos de Bactérias/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/microbiologia , Proliferação de Células , Células Dendríticas/citologia , Inflamação , Cinética , Linfonodos/citologia , Linfonodos/patologia , Ativação Linfocitária/imunologia , Camundongos , Fagócitos/microbiologia , Receptores CCR6 , Receptores de Quimiocinas/deficiência , Salmonella , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA