Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Immunol ; 50(9): 1295-1306, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32277709

RESUMO

Human IgA could be from different isotypes (IgA1/IgA2) and/or isoforms (monomeric, dimeric, or secretory). Monomeric IgA mainly IgA1 are considered as an anti-inflammatory isotype whereas dimeric/secretory IgA have clearly dual pro- and anti-inflammatory effects. Here, we show that IgA isotypes and isoforms display different binding abilities to FcαRI, Dectin-1, DC-SIGN, and CD71 on monocyte-derived dendritic cells (moDC). We describe that IgA regulate the expression of their own receptors and trigger modulation of moDC maturation. We also demonstrate that dimeric IgA2 and IgA1 induce different inflammatory responses leading to cytotoxic CD8+ T cells activation. moDC stimulation by dimeric IgA2 was followed by a strong pro-inflammatory effect. Our study highlights differences regarding IgA isotypes and isoforms in the context of DC conditioning. Further investigations are needed on the activation of adaptive immunity by IgA in the context of microbiota/IgA complexes during antibody-mediated immune selection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Imunoglobulina A/imunologia , Ativação Linfocitária/imunologia , Humanos , Imunoglobulina A/química , Isoformas de Proteínas
2.
J Virol ; 92(23)2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30232191

RESUMO

Noroviruses are enteric pathogens causing significant morbidity, mortality, and economic losses worldwide. Secretory immunoglobulins (sIg) are a first line of mucosal defense against enteric pathogens. They are secreted into the intestinal lumen via the polymeric immunoglobulin receptor (pIgR), where they bind to antigens. However, whether natural sIg protect against norovirus infection remains unknown. To determine if natural sIg alter murine norovirus (MNV) pathogenesis, we infected pIgR knockout (KO) mice, which lack sIg in mucosal secretions. Acute MNV infection was significantly reduced in pIgR KO mice compared to controls, despite increased MNV target cells in the Peyer's patch. Natural sIg did not alter MNV binding to the follicle-associated epithelium (FAE) or crossing of the FAE into the lymphoid follicle. Instead, naive pIgR KO mice had enhanced levels of the antiviral inflammatory molecules interferon gamma (IFN-γ) and inducible nitric oxide synthase (iNOS) in the ileum compared to controls. Strikingly, depletion of the intestinal microbiota in pIgR KO and control mice resulted in comparable IFN-γ and iNOS levels, as well as MNV infectious titers. IFN-γ treatment of wild-type (WT) mice and neutralization of IFN-γ in pIgR KO mice modulated MNV titers, implicating the antiviral cytokine in the phenotype. Reduced gastrointestinal infection in pIgR KO mice was also observed with another enteric virus, reovirus. Collectively, our findings suggest that natural sIg are not protective during enteric virus infection, but rather, that sIg promote enteric viral infection through alterations in microbial immune responses.IMPORTANCE Enteric virus, such as norovirus, infections cause significant morbidity and mortality worldwide. However, direct antiviral infection prevention strategies are limited. Blocking host entry and initiation of infection provides an established avenue for intervention. Here, we investigated the role of the polymeric immunoglobulin receptor (pIgR)-secretory immunoglobulin (sIg) cycle during enteric virus infections. The innate immune functions of sIg (agglutination, immune exclusion, neutralization, and expulsion) were not required during control of acute murine norovirus (MNV) infection. Instead, lack of pIgR resulted in increased IFN-γ levels, which contributed to reduced MNV titers. Another enteric virus, reovirus, also showed decreased infection in pIgR KO mice. Collectively, our data point to a model in which sIg-mediated microbial sensing promotes norovirus and reovirus infection. These data provide the first evidence of the proviral role of natural sIg during enteric virus infections and provide another example of how intestinal bacterial communities indirectly influence MNV pathogenesis.


Assuntos
Infecções por Caliciviridae/virologia , Trato Gastrointestinal/virologia , Imunoglobulinas/metabolismo , Receptores de Imunoglobulina Polimérica/fisiologia , Infecções por Reoviridae/virologia , Replicação Viral/imunologia , Animais , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/metabolismo , Trato Gastrointestinal/imunologia , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/metabolismo , Norovirus/imunologia , Reoviridae/imunologia , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/metabolismo
3.
J Allergy Clin Immunol ; 137(1): 214-222.e2, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26414879

RESUMO

BACKGROUND: Transmission of mucosal pathogens relies on their ability to bind to the surfaces of epithelial cells, to cross this thin barrier, and to gain access to target cells and tissues, leading to systemic infection. This implies that pathogen-specific immunity at mucosal sites is critical for the control of infectious agents using these routes to enter the body. Although mucosal delivery would ensure the best onset of protective immunity, most of the candidate vaccines are administered through the parenteral route. OBJECTIVE: The present study evaluates the feasibility of delivering the chemically bound p24gag (referred to as p24 in the text) HIV antigen through secretory IgA (SIgA) in nasal mucosae in mice. RESULTS: We show that SIgA interacts specifically with mucosal microfold cells present in the nasal-associated lymphoid tissue. p24-SIgA complexes are quickly taken up in the nasal cavity and selectively engulfed by mucosal dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin-positive dendritic cells. Nasal immunization with p24-SIgA elicits both a strong humoral and cellular immune response against p24 at the systemic and mucosal levels. This ensures effective protection against intranasal challenge with recombinant vaccinia virus encoding p24. CONCLUSION: This study represents the first example that underscores the remarkable potential of SIgA to serve as a carrier for a protein antigen in a mucosal vaccine approach targeting the nasal environment.


Assuntos
Proteína do Núcleo p24 do HIV/imunologia , Imunização/métodos , Imunoglobulina A Secretora/imunologia , Tecido Linfoide/imunologia , Mucosa Nasal/imunologia , Administração Intranasal , Animais , Citocinas/imunologia , Células Dendríticas , Feminino , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Tecido Linfoide/citologia , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucosa Nasal/citologia , Baço/imunologia , Vagina/imunologia
4.
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
5.
Eur J Immunol ; 45(3): 773-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25412898

RESUMO

HIV transmission and spread in the host are based on the survival of the virus or infected cells present in mucosal secretions, and the virus' ability to cross the epithelial barrier and access immune target cells, which leads to systemic infection. Therefore, HIV-specific immunity at mucosal sites is critical for control of infection. Although mucosal delivery would ensure the best onset of protective immunity, most candidate vaccines are administered through the parenteral route. Remarkably, secretory IgA (SIgA) interacts specifically with mucosal microfold (M) cells present in gut-associated lymphoid tissues. Here we evaluate the feasibility of delivering chemically bound p24HIV antigen via SIgA into the intestinal mucosae in mice. After oral administration, p24-SIgA complexes are quickly delivered into the tissue and selectively captured by CX3CR1(+) dendritic cells. Oral immunization with p24gag linked to SIgA (p24-SIgA) adjuvanted with E. coli heat labile enterotoxin (HLT) elicits both humoral and cellular immune responses against p24 at the systemic and mucosal levels and induces efficient protection against rectal challenge with a recombinant vaccinia virus encoding gag. This is the first study which underscores the remarkable potential of SIgA to serve as a vaccine carrier for an HIV antigen in mucosal administration targeting the gastrointestinal environment.


Assuntos
Vacinas contra a AIDS/farmacologia , Portadores de Fármacos/farmacologia , Proteína do Núcleo p24 do HIV/farmacologia , HIV-1/imunologia , Imunoglobulina A Secretora/farmacologia , Mucosa Intestinal/imunologia , Vacinas contra a AIDS/imunologia , Animais , Proteína do Núcleo p24 do HIV/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunoglobulina A Secretora/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
6.
PLoS Biol ; 11(9): e1001658, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24068891

RESUMO

Intestinal microfold (M) cells possess a high transcytosis capacity and are able to transport a broad range of materials including particulate antigens, soluble macromolecules, and pathogens from the intestinal lumen to inductive sites of the mucosal immune system. M cells are also the primary pathway for delivery of secretory IgA (SIgA) to the gut-associated lymphoid tissue. However, although the consequences of SIgA uptake by M cells are now well known and described, the mechanisms whereby SIgA is selectively bound and taken up remain poorly understood. Here we first demonstrate that both the Cα1 region and glycosylation, more particularly sialic acid residues, are involved in M cell-mediated reverse transcytosis. Second, we found that SIgA is taken up by M cells via the Dectin-1 receptor, with the possible involvement of Siglec-5 acting as a co-receptor. Third, we establish that transcytosed SIgA is taken up by mucosal CX3CR1⁺ dendritic cells (DCs) via the DC-SIGN receptor. Fourth, we show that mucosal and systemic antibody responses against the HIV p24-SIgA complexes administered orally is strictly dependent on the expression of Dectin-1. Having deciphered the mechanisms leading to specific targeting of SIgA-based Ag complexes paves the way to the use of such a vehicle for mucosal vaccination against various infectious diseases.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Imunoglobulina A Secretora/imunologia , Mucosa Intestinal/imunologia , Lectinas Tipo C/metabolismo , Transcitose/imunologia , Animais , Complexo Antígeno-Anticorpo/imunologia , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Células CHO , Receptor 1 de Quimiocina CX3C , Células CACO-2 , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Cricetulus , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Glicosilação , Proteína do Núcleo p24 do HIV/imunologia , Células HeLa , Humanos , Imunoglobulina A/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Intestinos/citologia , Lectinas/metabolismo , Lectinas Tipo C/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácido N-Acetilneuramínico/química , Receptores de Superfície Celular/metabolismo , Receptores de Quimiocinas/metabolismo
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.
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
10.
J Biol Chem ; 287(47): 40074-82, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23027876

RESUMO

The mammalian gastrointestinal (GI) tract harbors a diverse population of commensal species collectively known as the microbiota, which interact continuously with the host. From very early in life, secretory IgA (SIgA) is found in association with intestinal bacteria. It is considered that this helps to ensure self-limiting growth of the microbiota and hence participates in symbiosis. However, the importance of this association in contributing to the mechanisms ensuring natural host-microorganism communication is in need of further investigation. In the present work, we examined the possible role of SIgA in the transport of commensal bacteria across the GI epithelium. Using an intestinal loop mouse model and fluorescently labeled bacteria, we found that entry of commensal bacteria in Peyer's patches (PP) via the M cell pathway was mediated by their association with SIgA. Preassociation of bacteria with nonspecific SIgA increased their dynamics of entry and restored the reduced transport observed in germ-free mice known to have a marked reduction in intestinal SIgA production. Selective SIgA-mediated targeting of bacteria is restricted to the tolerogenic CD11c(+)CD11b(+)CD8(-) dendritic cell subset located in the subepithelial dome region of PPs, confirming that the host is not ignorant of its resident commensals. In conclusion, our work supports the concept that SIgA-mediated monitoring of commensal bacteria targeting dendritic cells in the subepithelial dome region of PPs represents a mechanism whereby the host mucosal immune system controls the continuous dialogue between the host and commensal bacteria.


Assuntos
Bactérias/imunologia , Células Dendríticas , Imunoglobulina A Secretora/imunologia , Mucosa Intestinal , Nódulos Linfáticos Agregados , Animais , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Feminino , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia
11.
Infect Immun ; 81(8): 3027-34, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23753631

RESUMO

Shigella flexneri, by invading intestinal epithelial cells (IECs) and inducing inflammatory responses of the colonic mucosa, causes bacillary dysentery. Although M cells overlying Peyer's patches are commonly considered the primary site of entry of S. flexneri, indirect evidence suggests that bacteria can also use IECs as a portal of entry to the lamina propria. Passive delivery of secretory IgA (SIgA), the major immunoglobulin secreted at mucosal surfaces, has been shown to protect rabbits from experimental shigellosis, but no information exists as to its molecular role in maintaining luminal epithelial integrity. We have established that the interaction of virulent S. flexneri with the apical pole of a model intestinal epithelium consisting of polarized Caco-2 cell monolayers resulted in the progressive disruption of the tight junction network and actin depolymerization, eventually resulting in cell death. The lipopolysaccharide (LPS)-specific agglutinating SIgAC5 monoclonal antibody (MAb), but not monomeric IgAC5 or IgGC20 MAbs of the same specificity, achieved protective functions through combined mechanisms, including limitation of the interaction between S. flexneri and epithelial cells, maintenance of the tight junction seal, preservation of the cell morphology, reduction of NF-κB nuclear translocation, and inhibition of proinflammatory mediator secretion. Our results add to the understanding of the function of SIgA-mediated immune exclusion by identifying a mode of action whereby the formation of immune complexes translates into maintenance of the integrity of epithelial cells lining the mucosa. This novel mechanism of protection mediated by SIgA is important to extend the arsenal of effective strategies to fight against S. flexneri mucosal invasion.


Assuntos
Disenteria Bacilar/imunologia , Imunoglobulina A Secretora/imunologia , Mucosa Intestinal/imunologia , Shigella flexneri/imunologia , Células CACO-2 , Ensaio de Imunoadsorção Enzimática , Humanos , Microscopia Eletrônica de Varredura , Shigella flexneri/patogenicidade , Junções Íntimas/imunologia
12.
Proc Natl Acad Sci U S A ; 107(7): 3064-9, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20133609

RESUMO

Sequentially along B cell differentiation, the different classes of membrane Ig heavy chains associate with the Ig alpha/Ig beta heterodimer within the B cell receptor (BCR). Whether each Ig class conveys specific signals adapted to the corresponding differentiation stage remains debated. We investigated the impact of the forced expression of an IgA-class receptor throughout murine B cell differentiation by knocking in the human C alpha Ig gene in place of the S mu region. Despite expression of a functional BCR, homozygous mutant mice showed a partial developmental blockade at the pro-B/pre-BI and large pre-BII cell stages, with decreased numbers of small pre-BII cells. Beyond this stage, peripheral B cell compartments of reduced size developed and allowed specific antibody responses, whereas mature cells showed constitutive activation and a strong commitment to plasma cell differentiation. Secreted IgA correctly assembled into polymers, associated with the murine J chain, and was transported into secretions. In heterozygous mutants, cells expressing the IgA allele competed poorly with those expressing IgM from the wild-type allele and were almost undetectable among peripheral B lymphocytes, notably in gut-associated lymphoid tissues. Our data indicate that the IgM BCR is more efficient in driving early B cell education and in mucosal site targeting, whereas the IgA BCR appears particularly suited to promoting activation and differentiation of effector plasma cells.


Assuntos
Linfócitos B/citologia , Cadeias alfa de Imunoglobulina/metabolismo , Cadeias mu de Imunoglobulina/metabolismo , Linfopoese/imunologia , Plasmócitos/citologia , Receptores de Antígenos de Linfócitos B/metabolismo , Animais , Linfócitos B/imunologia , Bromodesoxiuridina , Citometria de Fluxo , Marcação de Genes , Humanos , Cadeias alfa de Imunoglobulina/imunologia , Cadeias mu de Imunoglobulina/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Plasmócitos/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Biol Chem ; 286(19): 17239-47, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21454510

RESUMO

Humans live in symbiosis with 10(14) commensal bacteria among which >99% resides in their gastrointestinal tract. The molecular bases pertaining to the interaction between mucosal secretory IgA (SIgA) and bacteria residing in the intestine are not known. Previous studies have demonstrated that commensals are naturally coated by SIgA in the gut lumen. Thus, understanding how natural SIgA interacts with commensal bacteria can provide new clues on its multiple functions at mucosal surfaces. Using fluorescently labeled, nonspecific SIgA or secretory component (SC), we visualized by confocal microscopy the interaction with various commensal bacteria, including Lactobacillus, Bifidobacteria, Escherichia coli, and Bacteroides strains. These experiments revealed that the interaction between SIgA and commensal bacteria involves Fab- and Fc-independent structural motifs, featuring SC as a crucial partner. Removal of glycans present on free SC or bound in SIgA resulted in a drastic drop in the interaction with gram-positive bacteria, indicating the essential role of carbohydrates in the process. In contrast, poor binding of gram-positive bacteria by control IgG was observed. The interaction with gram-negative bacteria was preserved whatever the molecular form of protein partner used, suggesting the involvement of different binding motifs. Purified SIgA and SC from either mouse hybridoma cells or human colostrum exhibited identical patterns of recognition for gram-positive bacteria, emphasizing conserved plasticity between species. Thus, sugar-mediated binding of commensals by SIgA highlights the currently underappreciated role of glycans in mediating the interaction between a highly diverse microbiota and the mucosal immune system.


Assuntos
Carboidratos/química , Colostro/imunologia , Bactérias Gram-Positivas/metabolismo , Hibridomas/metabolismo , Imunoglobulina A Secretora/metabolismo , Intestinos/microbiologia , Motivos de Aminoácidos , Colostro/metabolismo , Colostro/microbiologia , Dimerização , Glicoproteínas/química , Glicosilação , Humanos , Hibridomas/microbiologia , Fragmentos de Imunoglobulinas/química , Lactobacillus/metabolismo , Microscopia Confocal/métodos , Polissacarídeos/química
14.
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
15.
J Biol Chem ; 285(2): 953-60, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19910466

RESUMO

In intestinal secretions, secretory IgA (SIgA) plays an important sentinel and protective role in the recognition and clearance of enteric pathogens. In addition to serving as a first line of defense, SIgA and SIgA x antigen immune complexes are selectively transported across Peyer's patches to underlying dendritic cells in the mucosa-associated lymphoid tissue, contributing to immune surveillance and immunomodulation. To explain the unexpected transport of immune complexes in face of the large excess of free SIgA in secretions, we postulated that SIgA experiences structural modifications upon antigen binding. To address this issue, we associated specific polymeric IgA and SIgA with antigens of various sizes and complexity (protein toxin, virus, bacterium). Compared with free antibody, we found modified sensitivity of the three antigens assayed after exposure to proteases from intestinal washes. Antigen binding further impacted on the immunoreactivity toward polyclonal antisera specific for the heavy and light chains of the antibody, as a function of the antigen size. These conformational changes promoted binding of the SIgA-based immune complex compared with the free antibody to cellular receptors (Fc alphaRI and polymeric immunoglobulin receptor) expressed on the surface of premyelocytic and epithelial cell lines. These data reveal that antigen recognition by SIgA triggers structural changes that confer to the antibody enhanced receptor binding properties. This identifies immune complexes as particular structural entities integrating the presence of bound antigens and adds to the known function of immune exclusion and mucus anchoring by SIgA.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Antígenos CD/imunologia , Imunoglobulina A/imunologia , Peptídeo Hidrolases/imunologia , Nódulos Linfáticos Agregados/imunologia , Receptores Fc/imunologia , Receptores de Imunoglobulina Polimérica/imunologia , Animais , Complexo Antígeno-Anticorpo/metabolismo , Antígenos/imunologia , Antígenos/metabolismo , Antígenos CD/metabolismo , Transporte Biológico/imunologia , Imunoglobulina A/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Peptídeo Hidrolases/metabolismo , Nódulos Linfáticos Agregados/enzimologia , Receptores Fc/metabolismo , Receptores de Imunoglobulina Polimérica/metabolismo
16.
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
17.
J Immunol ; 183(9): 5879-85, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19828639

RESUMO

Shigella, a Gram-negative invasive enteropathogenic bacterium responsible for bacillary dysentery, causes the rupture, invasion, and inflammatory destruction of the human colonic mucosa. We explored the mechanisms of protection mediated by Shigella LPS-specific secretory IgA (SIgA), the major mucosal Ab induced upon natural infection. Bacteria, SIgA, or SIgA-S. flexneri immune complexes were administered into rabbit ligated intestinal loops containing a Peyer's patch. After 8 h, localizations of bacteria, SIgA, and SIgA-S. flexneri immune complexes were examined by immunohistochemistry and confocal microscopy imaging. We found that anti-Shigella LPS SIgA, mainly via immune exclusion, prevented Shigella-induced inflammation responsible for the destruction of the intestinal barrier. Besides this luminal trapping, a small proportion of SIgA-S. flexneri immune complexes were shown to enter the rabbit Peyer's patch and were internalized by dendritic cells of the subepithelial dome region. Local inflammatory status was analyzed by quantitative RT-PCR using newly designed primers for rabbit pro- and anti-inflammatory mediator genes. In Peyer's patches exposed to immune complexes, limited up-regulation of the expression of proinflammatory genes, including TNF-alpha, IL-6, Cox-2, and IFN-gamma, was observed, consistent with preserved morphology. In contrast, in Peyer's patches exposed to Shigella alone, high expression of the same mediators was measured, indicating that neutralizing SIgA dampens the proinflammatory properties of Shigella. These results show that in the form of immune complexes, SIgA guarantees both immune exclusion and neutralization of translocated bacteria, thus preserving the intestinal barrier integrity by preventing bacterial-induced inflammation. These findings add to the multiple facets of the noninflammatory properties of SIgA.


Assuntos
Regulação para Baixo/imunologia , Disenteria Bacilar/imunologia , Disenteria Bacilar/patologia , Imunoglobulina A Secretora/fisiologia , Mediadores da Inflamação/antagonistas & inibidores , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Shigella flexneri/imunologia , Animais , Especificidade de Anticorpos , Permeabilidade da Membrana Celular/imunologia , Modelos Animais de Doenças , Disenteria Bacilar/prevenção & controle , Humanos , Íleo/imunologia , Íleo/metabolismo , Íleo/microbiologia , Íleo/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/antagonistas & inibidores , Coelhos , Shigella flexneri/crescimento & desenvolvimento
18.
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
19.
Cell Rep ; 37(7): 110006, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34788614

RESUMO

Secretory immunoglobulin A (SIgA) can travel to and from the lumen and transport antigen to subepithelial cells. However, IgM can also multimerize into functional secretory component-bound immunoglobulin. While it is already known that both SIgA and SIgM undergo transcytosis to be secreted at the mucosal surface, only SIgA has been shown to perform retrotranscytosis through microfold cells (M cells) of the Peyer's patch. Here, we investigate whether SIgM could also be taken up by M cells via retrotranscytosis. This transport involves FcµR binding at the apical membrane of M cells. We then demonstrate that SIgM can be exploited by SIgM-p24 (HIV-capsid protein) complexes during immunization in the nasal- or gut-associated lymphoid tissue (NALT or GALT), conferring efficient immune responses against p24. Our data demonstrate a mucosal function of SIgM, which could play a role in the regulation of mucosal immunity.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Intestinos/fisiologia , Proteínas de Membrana/metabolismo , Transcitose/fisiologia , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Feminino , Imunidade nas Mucosas/fisiologia , Imunoglobulina A Secretora/metabolismo , Imunoglobulina M/imunologia , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Nódulos Linfáticos Agregados/citologia , Transcitose/genética
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA