Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 261, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33431850

RESUMO

Intestinal microfold cells are the primary pathway for translocation of secretory IgA (SIgA)-pathogen complexes to gut-associated lymphoid tissue. Uptake of SIgA/commensals complexes is important for priming adaptive immunity in the mucosa. This study aims to explore the effect of SIgA retrograde transport of immune complexes in Crohn's disease (CD). Here we report a significant increase of SIgA transport in CD patients with NOD2-mutation compared to CD patients without NOD2 mutation and/or healthy individuals. NOD2 has an effect in the IgA transport through human and mouse M cells by downregulating Dectin-1 and Siglec-5 expression, two receptors involved in retrograde transport. These findings define a mechanism of NOD2-mediated regulation of mucosal responses to intestinal microbiota, which is involved in CD intestinal inflammation and dysbiosis.


Assuntos
Doença de Crohn/metabolismo , Imunoglobulina A Secretora/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Animais , Colite/microbiologia , Colite/patologia , Doença de Crohn/patologia , Humanos , Lectinas Tipo C/metabolismo , Camundongos Knockout , Modelos Biológicos , Mutação/genética , Proteína Adaptadora de Sinalização NOD2/deficiência , Proteína Adaptadora de Sinalização NOD2/genética , Nódulos Linfáticos Agregados/metabolismo , Transporte Proteico , Salmonella/fisiologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/metabolismo , Transcitose
2.
Artigo em Inglês | MEDLINE | ID: mdl-29423380

RESUMO

To define the bottlenecks that restrict antigen expression after oral administration of viral-vectored vaccines, we tracked vectors derived from the human adenovirus type 5 at whole body, tissue, and cellular scales throughout the digestive tract in a murine model of oral delivery. After intragastric administration of vectors encoding firefly luciferase or a model antigen, detectable levels of transgene-encoded protein or mRNA were confined to the intestine, and restricted to delimited anatomical zones. Expression of luciferase in the form of multiple small bioluminescent foci in the distal ileum, cecum, and proximal colon suggested multiple crossing points. Many foci were unassociated with visible Peyer's patches, implying that transduced cells lay in proximity to villous rather than follicle-associated epithelium, as supported by detection of transgene-encoded antigen in villous epithelial cells. Transgene-encoded mRNA but not protein was readily detected in Peyer's patches, suggesting that post-transcriptional regulation of viral gene expression might limit expression of transgene-encoded antigen in this tissue. To characterize the pathways by which the vector crossed the intestinal epithelium and encountered sentinel cells, a fluorescent-labeled vector was administered to mice by the intragastric route or inoculated into ligated intestinal loops comprising a Peyer's patch. The vector adhered selectively to microfold cells in the follicle-associated epithelium, and, after translocation to the subepithelial dome region, was captured by phagocytes that expressed CD11c and lysozyme. In conclusion, although a large number of crossing events took place throughout the intestine within and without Peyer's patches, multiple firewalls prevented systemic dissemination of vector and suppressed production of transgene-encoded protein in Peyer's patches.


Assuntos
Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Transgenes/genética , Transgenes/imunologia , Administração Oral , Animais , Feminino , Expressão Gênica , Genes Reporter , Vetores Genéticos/administração & dosagem , Humanos , Imunização , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos , Especificidade de Órgãos , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Fagócitos/metabolismo , Transporte Proteico , Vacinação
3.
Gut Microbes ; 8(5): 511-519, 2017 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-28541767

RESUMO

Despite important success in protecting individuals against many pathogenic infections, parenteral vaccination is not optimal to induce immunity at the site of pathogen entry, i.e. mucosal surfaces. Moreover, designing adequate delivery systems and safe adjuvants to overcome the inherent tolerogenic environment of the mucosal tissue is challenging, in particular in the gastrointestinal tract prone to antigen degradation. We recently demonstrated that intranasal administration of a Salmonella-derived antigen associated with gas-filled microbubbles induced specific Ab and T cell responses in the gut and was associated with a reduction in local and systemic bacterial load after oral Salmonella infection. Building on these promising data, the adequate choice of antigen(s) to be administered and how to make it suitable for possible human application are discussed. We additionally present novel data dealing with oral administration of microbubbles and describe research strategies to direct them to mucosal sampling/inductive sites.


Assuntos
Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Portadores de Fármacos/administração & dosagem , Sistemas de Liberação de Medicamentos , Trato Gastrointestinal/imunologia , Microbolhas , Salmonelose Animal/prevenção & controle , Administração através da Mucosa , Animais , Anticorpos Antibacterianos/análise , Carga Bacteriana , Imunidade nas Mucosas , Camundongos , Salmonella/isolamento & purificação , Salmonelose Animal/imunologia , Linfócitos T/imunologia
4.
Int J Pharm ; 514(1): 176-188, 2016 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-27863662

RESUMO

Biodegradable poly(lactic-co-glycolic acid) (PLGA) microspheres are potential vehicles to deliver antigens for vaccination. Because they lack the full capacity to activate professional antigen presenting cells (APCs), combination with an immunostimulatory adjuvant may be considered. A candidate is the synthetic TLR3 ligand polyriboinosinic acid-polyribocytidylic acid, poly(I:C), which drives cell-mediated immunity. However, poly(I:C) has also been linked to the pathogenesis of autoimmunity, as affected by widespread stimulation of non-hematopoietic bystander cells. To address this aspect, we propose to minimize the poly(I:C) dose as well as to control the stimulation of non-immune bystander cells by poly(I:C). To facilitate the maturation of APCs with minimal poly(I:C) doses, we surface-assembled poly(I:C) onto PLGA microspheres. The microspheres' surface was further modified by poly(ethylene glycol) (PEG) coronas with varying PEG-densities. PLGA microspheres loaded with tetanus toxoid (tt) as model antigen were manufactured by microextrusion-based solvent extraction. The negatively charged PLGA(tt) microspheres were coated with polycationic poly(l-lysine) (PLL) polymers, either PLL itself or PEG-grafted PLL (PLL-g-PEG) with varying grafting ratios (g=2.2 and g=10.1). Stable surface assembly of poly(I:C) was achieved by subsequent incubation of polymer-coated PLGA microspheres with aqueous poly(I:C) solutions. We evaluated the immunostimulatory potential of such PLGA(tt) microsphere formulations on monocyte-derived dendritic cells (MoDCs) as well as human foreskin fibroblasts (HFFs) as model for non-hematopoietic bystander cells. Formulations with surface-assembled poly(I:C) readily activated MoDCs with respect to the expression of maturation-related surface markers, proinflammatory cytokine secretion and directed migration. When surface-assembled, poly(I:C) enhanced its immunostimulatory activity by more than one order of magnitude as compared to free poly(I:C). On fibroblasts, surface-assembled poly(I:C) upregulated class I MHC but not class II MHC. Phagocytosis of PLGA(tt) microsphere formulations by MoDCs and HFFs remained mostly unaffected by PEG-grafted PLL coatings. In contrast, high concentrations of free poly(I:C) led to a marked drop of microsphere phagocytosis by HFFs. Overall, surface assembly on PEGylated PLGA microspheres holds promise to improve both efficacy and safety of poly(I:C) as vaccine adjuvant.


Assuntos
Adjuvantes Imunológicos/química , Antígenos/química , Ácido Láctico/química , Poli I-C/química , Polietilenoglicóis/química , Ácido Poliglicólico/química , Polilisina/análogos & derivados , Vacinas/química , Adjuvantes Imunológicos/farmacologia , Antígenos/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Fibroblastos/imunologia , Humanos , Imunidade Celular/imunologia , Microesferas , Monócitos/imunologia , Fagocitose/imunologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polilisina/química , Propriedades de Superfície , Toxoide Tetânico/química , Toxoide Tetânico/imunologia , Vacinas/imunologia
5.
J Infect Dis ; 214(3): 438-46, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27122591

RESUMO

Salmonella infection is an increasingly important public health problem owing to the emergence of multidrug resistance and the lack of broadly efficient vaccines. Novel strategies of vaccination are required to induce protective immune responses at mucosal surfaces and in the circulation, to limit bacteria entry and dissemination. To this aim, intranasal anti-Salmonella vaccination with an innovative formulation composed of gas-filled microbubbles and the pathogen-derived protective protein serodominant secreted effector protein B (SseB-MB) was evaluated in a mouse infection model. Intranasal application of SseB-MB induced gut and systemic immunoglobulin A, T-helper type 17 cell (Th17), and Th1 responses, all of which are associated with natural immunity against Salmonella In vaccinated mice, a significant reduction in bacterial load was observed in intestinal tissues and the spleen after an otherwise lethal oral infection. Therefore, MB serve as an efficient carrier for nasal delivery of a Salmonella antigen that results in protection upon activation of the common mucosal immune system.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Portadores de Fármacos/administração & dosagem , Trato Gastrointestinal/imunologia , Chaperonas Moleculares/imunologia , Infecções por Salmonella/prevenção & controle , Vacinas contra Salmonella/imunologia , Salmonella/imunologia , Administração Intranasal , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/administração & dosagem , Carga Bacteriana , Proteínas de Bactérias/administração & dosagem , Modelos Animais de Doenças , Feminino , Trato Gastrointestinal/microbiologia , Imunidade nas Mucosas , Imunoglobulina A/análise , Imunoglobulina A/sangue , Camundongos Endogâmicos BALB C , Chaperonas Moleculares/administração & dosagem , Infecções por Salmonella/imunologia , Vacinas contra Salmonella/administração & dosagem , Células Th1/imunologia , Células Th17/imunologia , Resultado do Tratamento
6.
Biomaterials ; 57: 153-60, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25916503

RESUMO

Vaccination aims at generating memory immune responses able to protect individuals against pathogenic challenges over long periods of time. Subunit vaccine formulations based on safe, but poorly immunogenic, antigenic entities must be combined with adjuvant molecules to make them efficient against infections. We have previously shown that gas-filled microbubbles (MB) are potent antigen-delivery systems. This study compares the ability of various ovalbumin-associated MB (OVA-MB) formulations to induce antigen-specific memory immune responses and evaluates long-term protection toward bacterial infections. When initially testing dendritic cells reactivity to MB constituents, palmitic acid exhibited the highest degree of activation. Subcutaneous immunization of naïve wild-type mice with the OVA-MB formulation comprising the highest palmitic acid content and devoid of PEG2000 was found to trigger the more pronounced Th1-type response, as reflected by robust IFN-γ and IL-2 production. Both T cell and antibody responses persisted for at least 6 months after immunization. At that time, systemic infection with OVA-expressing Listeria monocytgenes was performed. Partial protection of vaccinated mice was demonstrated by reduction of the bacterial load in both the spleen and liver. We conclude that antigen-bound MB exhibit promising properties as a vaccine candidate ensuring prolonged maintenance of protective immunity.


Assuntos
Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/uso terapêutico , Infecções por Bactérias Gram-Positivas/prevenção & controle , Listeria/imunologia , Microbolhas , Ovalbumina/administração & dosagem , Ovalbumina/uso terapêutico , Animais , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Feminino , Expressão Gênica , Infecções por Bactérias Gram-Positivas/imunologia , Humanos , Imunidade , Interferon gama/imunologia , Interleucina-2/imunologia , Listeria/genética , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/genética , Ovalbumina/imunologia , Recombinação Genética , Vacinação
7.
J Biol Chem ; 289(31): 21617-26, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24951593

RESUMO

Intravenous administration of polyclonal and monoclonal antibodies has proven to be a clinically valid approach in the treatment, or at least relief, of many acute and chronic pathologies, such as infection, immunodeficiency, and a broad range of autoimmune conditions. Plasma-derived IgG or recombinant IgG are most frequently used for intravenous or subcutaneous administration, whereas a few IgM-based products are available as well. We have established recently that secretory-like IgA and IgM can be produced upon association of plasma-derived polymeric IgA and IgM with a recombinant secretory component. As a next step toward potential future mucosal administration, we sought to unravel the mechanisms by which these secretory Igs protect epithelial cells located at the interface between the environment and the inside of the body. By using polarized epithelial Caco-2 cell monolayers and Shigella flexneri as a model enteropathogen, we found that polyspecific plasma-derived SIgA and SIgM fulfill many protective functions, including dose-dependent recognition of the antigen via formation of aggregated immune complexes, reduction of bacterial infectivity, maintenance of epithelial cell integrity, and inhibition of proinflammatory cytokine/chemokine production by epithelial cells. In this in vitro model devoid of other cellular or molecular interfering partners, IgM and secretory IgM showed stronger bacterial neutralization than secretory IgA. Together, these data suggest that mucosally delivered antibody preparations may be most effective when combining both secretory-like IgA and IgM, which, together, play a crucial role in preserving several levels of epithelial cell integrity.


Assuntos
Células Epiteliais/citologia , Imunoglobulina A/imunologia , Imunoglobulina M/imunologia , Shigella flexneri/fisiologia , Células CACO-2 , Quimiocinas/metabolismo , Contagem de Colônia Microbiana , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Homeostase , Humanos , Imunoglobulina A/sangue , Imunoglobulina M/sangue , Microscopia Confocal , Shigella flexneri/patogenicidade , Virulência
8.
J Immunol ; 191(5): 2335-43, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23926325

RESUMO

IgA plays ambivalent roles in the immune system. The balance between inhibitory and activating responses relies on the multimerization status of IgA and interaction with their cognate receptors. In mucosal sites, secretory IgA (SIgA) protects the host through immune-exclusion mechanisms, but its function in the bloodstream remains unknown. Using bone marrow-derived dendritic cells, we found that both human and mouse SIgA induce tolerogenic dendritic cells (DCs) following binding to specific ICAM-3 grabbing nonintegrin receptor 1. This interaction was dependent on Ca(2+) and mannose residues. SIgA-primed DCs (SIgA-DCs) are resistant to TLR-dependent maturation. Although SIgA-DCs fail to induce efficient proliferation and Th1 differentiation of naive responder T cells, they generate the expansion of regulatory T cells through IL-10 production. SIgA-DCs are highly potent in inhibiting autoimmune responses in mouse models of type 1 diabetes and multiple sclerosis. This discovery may offer new insights about mucosal-derived DC immunoregulation through SIgA opening new therapeutic approaches to autoimmune diseases.


Assuntos
Autoimunidade/imunologia , Moléculas de Adesão Celular/imunologia , Células Dendríticas/imunologia , Imunoglobulina A Secretora/imunologia , Lectinas Tipo C/imunologia , Receptores de Superfície Celular/imunologia , Animais , Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Tolerância Imunológica/imunologia , Camundongos , Camundongos Endogâmicos C57BL
9.
Adv Drug Deliv Rev ; 65(10): 1386-99, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23751781

RESUMO

Current research and development of antigens for vaccination often center on purified recombinant proteins, viral subunits, synthetic oligopeptides or oligosaccharides, most of them suffering from being poorly immunogenic and subject to degradation. Hence, they call for efficient delivery systems and potent immunostimulants, jointly denoted as adjuvants. Particulate delivery systems like emulsions, liposomes, nanoparticles and microspheres may provide protection from degradation and facilitate the co-formulation of both the antigen and the immunostimulant. Synthetic double-stranded (ds) RNA, such as polyriboinosinic acid-polyribocytidylic acid, poly(I:C), is a mimic of viral dsRNA and, as such, a promising immunostimulant candidate for vaccines directed against intracellular pathogens. Poly(I:C) signaling is primarily dependent on Toll-like receptor 3 (TLR3), and on melanoma differentiation-associated gene-5 (MDA-5), and strongly drives cell-mediated immunity and a potent type I interferon response. However, stability and toxicity issues so far prevented the clinical application of dsRNAs as they undergo rapid enzymatic degradation and bear the potential to trigger undue immune stimulation as well as autoimmune disorders. This review addresses these concerns and suggests strategies to improve the safety and efficacy of immunostimulatory dsRNA formulations. The focus is on technological means required to lower the necessary dosage of poly(I:C), to target surface-modified microspheres passively or actively to antigen-presenting cells (APCs), to control their interaction with non-professional phagocytes and to modulate the resulting cytokine secretion profile.


Assuntos
Adjuvantes Imunológicos/química , Poli I-C/química , Vacinas/química , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígenos/administração & dosagem , Antígenos/química , Células Dendríticas/imunologia , Humanos , Microesferas , Poli I-C/administração & dosagem , Receptor 3 Toll-Like/imunologia , Vacinas/administração & dosagem
10.
Biomaterials ; 34(21): 5423-30, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23618628

RESUMO

Gas-filled microbubbles (MB) are a very promising alternative to the currently evaluated lipid- or polymer-based particulate Ag delivery systems. We recently demonstrated the ability of MB to deliver associated Ag to DC, to activate them and thereby induce both humoral and cellular immune responses. We now extended the characterization of MB as antigen-delivery system by appraising the efficiency of MB-associated ovalbumin (OVA-MB) at protecting mice against pathogen infection. Ultrasound-mediated imaging demonstrated that the administration of OVA via MB generates a depot at the injection site that lasts for several hours. We found that OVA-MB injected subcutaneously is far more effective at inducing specific Ab and T cell immunity than immunization with free OVA. Moreover, a covalent link between MB and OVA causes a stronger bias towards a Th1-type of immune response than adsorption of the Ag or its covalent link to liposomes of the same lipid composition. Finally, vaccination of mice with OVA-MB partially protects against a systemic infection with OVA-expressing Listeria monocytogenes. The vaccine induces specific effector CD8 T cell responses capable of decreasing more than 100 fold the bacterial load. MB thus represent a potent Ag delivery system for vaccination against intracellular infectious agents.


Assuntos
Vacinas Bacterianas/imunologia , Gases/química , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/prevenção & controle , Microbolhas , Ovalbumina/imunologia , Animais , Formação de Anticorpos/imunologia , Feminino , Imunidade Humoral/imunologia , Injeções , Listeriose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Ultrassom , Vacinação
11.
J Biol Chem ; 288(6): 4085-94, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23250751

RESUMO

Immunotherapy with monoclonal and polyclonal immunoglobulin is successfully applied to improve many clinical conditions, including infection, autoimmune diseases, or immunodeficiency. Most immunoglobulin products, recombinant or plasma-derived, are based on IgG antibodies, whereas to date, the use of IgA for therapeutic application has remained anecdotal. In particular, purification or production of large quantities of secretory IgA (SIgA) for potential mucosal application has not been achieved. In this work, we sought to investigate whether polymeric IgA (pIgA) recovered from human plasma is able to associate with secretory component (SC) to generate SIgA-like molecules. We found that ∼15% of plasma pIgA carried J chain and displayed selective SC binding capacity either in a mixture with monomeric IgA (mIgA) or after purification. The recombinant SC associated covalently in a 1:1 stoichiometry with pIgA and with similar efficacy as colostrum-derived SC. In comparison with pIgA, the association with SC delayed degradation of SIgA by intestinal proteases. Similar results were obtained with plasma-derived IgM. In vitro, plasma-derived IgA and SIgA neutralized Shigella flexneri used as a model pathogen, resulting in a delay of bacteria-induced damage targeted to polarized Caco-2 cell monolayers. The sum of these novel data demonstrates that association of plasma-derived IgA or IgM with recombinant/colostrum-derived SC is feasible and yields SIgA- and SIgM-like molecules with similar biochemical and functional characteristics as mucosa-derived immunoglobulins.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/imunologia , Cadeias J de Imunoglobulina/imunologia , Imunoglobulina M/imunologia , Plasma/imunologia , Componente Secretório/imunologia , Shigella flexneri/imunologia , Animais , Anticorpos Antibacterianos/química , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/isolamento & purificação , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/isolamento & purificação , Células COS , Chlorocebus aethiops , Humanos , Cadeias J de Imunoglobulina/química , Cadeias J de Imunoglobulina/genética , Cadeias J de Imunoglobulina/isolamento & purificação , Imunoglobulina M/química , Imunoglobulina M/genética , Imunoglobulina M/isolamento & purificação , Plasma/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Componente Secretório/química , Componente Secretório/genética , Componente Secretório/isolamento & purificação
12.
Biomaterials ; 33(25): 5935-46, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22652023

RESUMO

The use of well characterized recombinant or purified protein antigens (Ag) for vaccination is of interest for safety reasons and in the case where inactivated pathogens are not available (cancer, allergy). However it requires the addition of adjuvants such as Ag carrier or immune stimulators to potentiate their immunogenicity. In this study, we demonstrated that gas-filled microbubbles (MB) can serve as an efficient Ag delivery system to promote phagocytosis of the model Ag ovalbumin (OVA) without the need of ultrasound application. Once internalized by DC, OVA was processed and presented to both CD4 and CD8 T cells in vitro; such observations were coupled with the capacity of MB to activate DC. In vivo administration of MB-associated OVA in naïve wild-type Balb/c mice resulted in the induction of OVA-specific antibody and T cell responses. Detailed characterization of the generated immune response demonstrated the production of both IgG1 and IgG2a serum antibodies, as well as the secretion of IFN-γ and IL-10 by splenocytes. Interestingly, similar results were obtained with human DC in regards of Ag delivery and cell activation. Therefore, the data presented here settle the proof of principle for the further evaluation of MB-based immunomodulation studies.


Assuntos
Antígenos/administração & dosagem , Antígenos/imunologia , Sistemas de Liberação de Medicamentos/métodos , Imunidade/imunologia , Microbolhas , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Animais , Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Fluorescência , Gases/química , Humanos , Hibridomas/imunologia , Injeções , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/imunologia , Fagocitose/imunologia
13.
Biomaterials ; 33(1): 333-42, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21983137

RESUMO

This study was designed to evaluate the potential of gas-filled microbubbles (MB) to be internalized by antigen-presenting cells (APC). Fluorescently labeled MB were prepared, thus permitting to track binding to, and internalization in, APC. Both human and mouse cells, including monocytes and dendritic cells (DC), prove capable to phagocyte MB in vitro. Observation by confocal laser scanning microscopy showed that interaction between MB and target cells resulted in a rapid internalization in cellular compartments and to a lesser extent in the cytoplasm. Capture of MB by APC resulted in phagolysosomal targeting as verified by double staining with anti-lysosome-associated membrane protein-1 monoclonal antibody and decrease of internalization by phagocytosis inhibitors. Fluorescent MB injected subcutaneously (s.c.) in mice were found to be associated with CD11c(+)DC in lymph nodes draining the injection sites 24 h after administration. Altogether, our study demonstrates that MB can successfully target APC both in vitro and in vivo, and thus may serve as a potent Ag delivery system without requirement for ultrasound-based sonoporation. This adds to the potential of applications of MB already extensively used for diagnostic imaging in humans.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Gases/química , Microbolhas , Fagocitose/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , Humanos , Camundongos
14.
Gut Microbes ; 2(5): 287-93, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22067937

RESUMO

Human beings live in symbiosis with billions of microorganisms colonizing mucosal surfaces. The understanding of the mechanisms underlying this fine-tuned intestinal balance has made significant processes during the last decades. We have recently demonstrated that the interaction of SIgA with Gram-positive bacteria is essentially based on Fab-independent, glycan-mediated recognition. Results obtained using mouse hybridoma- and colostrum-derived secretory IgA (SIgA) consistently show that N-glycans present on secretory component (SC) play a crucial role in the process. Natural coating may involve specific Gram-positive cell wall components, which may explain selective recognition at the molecular level. More widely, the existence of these complexes is involved in the modulation of intestinal epithelial cell (IEC) responses in vitro and the formation of intestinal biofilms. Thus, SIgA may act as one of the pillars in homeostatic maintenance of the microbiota in the gut, adding yet another facet to its multiple roles in the mucosal environment.


Assuntos
Bactérias Gram-Positivas/fisiologia , Imunoglobulina A Secretora/metabolismo , Intestinos/microbiologia , Polissacarídeos/metabolismo , Componente Secretório/metabolismo , Animais , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Bactérias Gram-Positivas/genética , Homeostase , Humanos , Imunoglobulina A Secretora/química , Intestinos/fisiologia , Camundongos , Ligação Proteica , Componente Secretório/química , Simbiose
15.
mBio ; 2(3): e00042-11, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21610121

RESUMO

UNLABELLED: Mucosal immunity to the enteric pathogen Shigella flexneri is mediated by secretory IgA (S-IgA) antibodies directed against the O-antigen (O-Ag) side chain of lipopolysaccharide. While secretory antibodies against the O-Ag are known to prevent bacterial invasion of the intestinal epithelium, the mechanisms by which this occurs are not fully understood. In this study, we report that the binding of a murine monoclonal IgA (IgAC5) to the O-Ag of S. flexneri serotype 5a suppresses activity of the type 3 secretion (T3S) system, which is necessary for S. flexneri to gain entry into intestinal epithelial cells. IgAC5's effects on the T3S were rapid (5 to 15 min) and were coincident with a partial reduction in the bacterial membrane potential and a decrease in intracellular ATP levels. Activity of the T3S system returned to normal levels 45 to 90 min following antibody treatment, demonstrating that IgAC5's effects were transient. Nonetheless, these data suggest a model in which the association of IgA with the O-Ag of S. flexneri partially de-energizes the T3S system and temporarily renders the bacterium incapable of invading intestinal epithelial cells. IMPORTANCE: Secretory IgA (S-IgA) serves as the first line of defense against enteric infections. However, despite its well-recognized role in mucosal immunity, relatively little is known at the molecular level about how this class of antibody functions to prevent pathogenic bacteria from penetrating the epithelial barrier. It is generally assumed that S-IgA functions primarily by "immune exclusion," a phenomenon in which the antibody binds to microbial surface antigens and thereby promotes bacterial agglutination, entrapment in mucus, and physical clearance from the gastrointestinal tract via peristalsis. The results of the present study suggest that in addition to serving as a physical barrier, S-IgA may have a direct impact on the ability of microbial pathogens to secrete virulence factors required for invasion of intestinal epithelial cells.


Assuntos
Anticorpos Antibacterianos/imunologia , Imunoglobulina A/imunologia , Proteínas de Membrana Transportadoras/metabolismo , Antígenos O/imunologia , Shigella flexneri/imunologia , Shigella flexneri/metabolismo , Fatores de Virulência/metabolismo , Trifosfato de Adenosina/análise , Animais , Anticorpos Antibacterianos/isolamento & purificação , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Proteínas de Bactérias/metabolismo , Membrana Celular/fisiologia , Citoplasma/química , Imunoglobulina A/isolamento & purificação , Potenciais da Membrana , Camundongos , Fatores de Tempo
16.
J Proteome Res ; 10(4): 1558-70, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21410266

RESUMO

Allergy is an immunological disorder of the upper airways, lung, skin, and the gut with a growing prevalence over the last decades in Western countries. Atopy, the genetic predisposition for allergy, is strongly dependent on familial inheritance and environmental factors. These observations call for predictive markers of progression from atopy to allergy, a prerequisite to any active intervention in neonates and children (prophylactic interventions/primary prevention) or in adults (immunomodulatory interventions/secondary prevention). In an attempt to identify early biomarkers of the "atopic march" using minimally invasive sampling, CD4+ T cells from 20 adult volunteers (10 healthy and 10 with respiratory allergies) were isolated and quantitatively analyzed and their proteomes were compared in and out of pollen season (± antigen exposure). The proteome study based on high-resolution 2D gel electrophoresis revealed three candidate protein markers that distinguish the CD4+ T cell proteomes of normal from allergic individuals when sampled out of pollen season, namely Talin 1, Nipsnap homologue 3A, and Glutamate-cysteine ligase regulatory protein. Three proteins were found differentially expressed between the CD4+ T cell proteomes of normal and allergic subjects when sampled during pollen season: carbonyl reductase, glutathione S-transferase ω 1, and 2,4-dienoyl-CoA reductase. The results were partly validated by Western blotting.


Assuntos
Alérgenos/imunologia , Biomarcadores/metabolismo , Linfócitos T CD4-Positivos/química , Linfócitos T CD4-Positivos/imunologia , Pólen/imunologia , Proteômica/métodos , Rinite Alérgica Sazonal/imunologia , Adulto , Feminino , Humanos , Hipersensibilidade/imunologia , Dados de Sequência Molecular , Proteoma/análise , Adulto Jovem
17.
J Biol Chem ; 285(44): 33906-13, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20729211

RESUMO

The precise mechanisms underlying the interaction between intestinal bacteria and the host epithelium lead to multiple consequences that remain poorly understood at the molecular level. Deciphering such events can provide valuable information as to the mode of action of commensal and probiotic microorganisms in the gastrointestinal environment. Potential roles of such microorganisms along the privileged target represented by the mucosal immune system include maturation prior, during and after weaning, and the reduction of inflammatory reactions in pathogenic conditions. Using human intestinal epithelial Caco-2 cell grown as polarized monolayers, we found that association of a Lactobacillus or a Bifidobacterium with nonspecific secretory IgA (SIgA) enhanced probiotic adhesion by a factor of 3.4-fold or more. Bacteria alone or in complex with SIgA reinforced transepithelial electrical resistance, a phenomenon coupled with increased phosphorylation of tight junction proteins zonula occludens-1 and occludin. In contrast, association with SIgA resulted in both enhanced level of nuclear translocation of NF-κB and production of epithelial polymeric Ig receptor as compared with bacteria alone. Moreover, thymic stromal lymphopoietin production was increased upon exposure to bacteria and further enhanced with SIgA-based complexes, whereas the level of pro-inflammatory epithelial cell mediators remained unaffected. Interestingly, SIgA-mediated potentiation of the Caco-2 cell responsiveness to the two probiotics tested involved Fab-independent interaction with the bacteria. These findings add to the multiple functions of SIgA and underscore a novel role of the antibody in interaction with intestinal bacteria.


Assuntos
Imunoglobulina A Secretora/química , Intestinos/citologia , Probióticos/química , Aderência Bacteriana , Bifidobacterium/metabolismo , Células CACO-2 , Células Epiteliais/citologia , Humanos , Lactobacillus/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Ocludina , Fosfoproteínas/metabolismo , Fosforilação , Junções Íntimas , Proteína da Zônula de Oclusão-1
18.
Int J Cancer ; 126(10): 2469-78, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19816937

RESUMO

Cervical cancer, the second leading cause of cancer mortality in women worldwide, results from infection with a subset of human papillomaviruses (HPV), HPV-16 being the most prevalent type. The available prophylactic vaccines are an effective strategy to prevent this cancer in the long term. However, they only target 70-80% of all cervical cancers and cannot control existing HPV infections and associated lesions. Therapeutic vaccines are thus necessary for women who cannot benefit from prophylactic vaccination. Induction of protective immune responses in the genital mucosa (GM) may be crucial for efficacy of HPV therapeutic vaccines. We report here that mice that received a single subcutaneous (s.c.) vaccination of an adjuvanted long synthetic HPV16 E7(1-98) polypeptide showed induction of 100% tumor protection against s.c. TC-1 tumors and that tumor regression was mainly provided by CD8 T cells. In vivo cytotoxic assay revealed high E7-specific cytolytic T lymphocytes activity in spleen and in genital draining lymph nodes (LN), and E7-specific CD8 T cells could be detected in GM by tetramer staining. More importantly, high-avidity E7-specific INF-gamma secreting CD8 T cells were induced not only in blood, spleen and LN but also in GM of vaccinated mice, thus providing evidence that a parenteral vaccination may be sufficient to provide regression of genital tumors. In addition, there was no correlation between the responses measured in blood with those measured in GM, highlighting the necessity and relevance to determine the immune responses in the mucosa where HPV-tumors reside.


Assuntos
Alphapapillomavirus/imunologia , Linfócitos T CD8-Positivos/imunologia , Genitália Feminina/imunologia , Infecções por Papillomavirus/tratamento farmacológico , Vacinas contra Papillomavirus/administração & dosagem , Infecções Tumorais por Vírus/tratamento farmacológico , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Feminino , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/imunologia , Proteínas E7 de Papillomavirus/imunologia , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/imunologia , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/imunologia , Neoplasias do Colo do Útero/imunologia , Neoplasias do Colo do Útero/virologia
19.
J Immunol ; 179(5): 3144-52, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17709529

RESUMO

Both IgG and secretory IgA Abs in mucosal secretions have been implicated in blocking the earliest events in HIV-1 transit across epithelial barriers, although the mechanisms by which this occurs remain largely unknown. In this study, we report the production and characterization of a human rIgA(2) mAb that carries the V regions of IgG1 b12, a potent and broadly neutralizing anti-gp120 Ab which has been shown to protect macaques against vaginal simian/HIV challenge. Monomeric, dimeric, polymeric, and secretory IgA(2) derivatives of b12 reacted with gp120 and neutralized CCR5- and CXCR4-tropic strains of HIV-1 in vitro. With respect to the protective effects of these Abs at mucosal surfaces, we demonstrated that IgG1 b12 and IgA(2) b12 inhibited the transfer of cell-free HIV-1 from ME-180 cells, a human cervical epithelial cell line, as well as Caco-2 cells, a human colonic epithelial cell line, to human PBMCs. Inhibition of viral transfer was due to the ability of b12 to block both viral attachment to and uptake by epithelial cells. These data demonstrate that IgG and IgA MAbs directed against a highly conserved epitope on gp120 can interfere with the earliest steps in HIV-1 transmission across mucosal surfaces, and reveal a possible mechanism by which b12 protects the vaginal mucosal against viral challenge in vivo.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , Imunoglobulina A Secretora/farmacologia , Imunoglobulina G/farmacologia , Região Variável de Imunoglobulina/farmacologia , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/genética , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Anticorpos Anti-HIV/biossíntese , Anticorpos Anti-HIV/genética , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/patogenicidade , Humanos , Imunoglobulina A Secretora/biossíntese , Imunoglobulina A Secretora/genética , Imunoglobulina G/biossíntese , Imunoglobulina G/genética , Região Variável de Imunoglobulina/biossíntese , Região Variável de Imunoglobulina/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
20.
J Biol Chem ; 281(20): 14280-7, 2006 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-16543244

RESUMO

In mucosal secretions, secretory component (SC) is found either free or bound to polymeric IgA within the secretory IgA complex. SC displays numerous and various glycans, which are potential ligands for bacterial compounds. We first established that human SC (hSC) purified from colostrum (hSCcol) or produced in Chinese hamster ovary cells (hSCrec) exhibits the same lectin reactivity. Both forms bind to Clostridium difficile toxin A and functionally protect polarized Caco-2 cell monolayers from the cytopathic effect of the toxin. The interaction is mediated by glycans present on hSC and involves galactose and sialic acid residues. hSCcol and hSCrec were also shown to bind enteropathogenic Escherichia coli adhesin intimin and to inhibit its infectivity on HEp-2 cells in a glycan-dependent manner as well. SC remained operative in the context of the whole secretory IgA molecule and can therefore enhance its Fab-mediated neutralizing properties. On the contrary, hSC did not interact with three different strains of rotavirus (RF, RRV, and SA11). Accordingly, infection of target MA104 cells with these rotavirus strains was not reduced in the presence of either form of hSC tested. Although not a universal mechanism, these findings identify hSC as a microbial scavenger contributing to the antipathogenic arsenal that protects the body epithelial surfaces.


Assuntos
Toxinas Bacterianas/química , Enterotoxinas/química , Mucosa/metabolismo , Polissacarídeos/química , Animais , Aderência Bacteriana , Células CHO , Células CACO-2 , Linhagem Celular Tumoral , Cricetinae , Escherichia coli/metabolismo , Humanos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Rotavirus/genética , Rotavirus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA