Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Vaccines (Basel) ; 11(3)2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36992257

RESUMO

RSV is divided into two antigenic subtypes, RSV A and RSV B, which is largely based on the variation in the G protein, while the fusion protein F is more conserved and a target for antibody-mediated neutralization. Here we evaluate the breadth of the protective immune responses across RSV A and RSV B subtypes, induced by vaccines based on the RSV A-based fusion protein, stabilized in the prefusion conformation (preF) in preclinical models. Immunization of naïve cotton rats with preF subunit or preF encoded by a replication incompetent Adenoviral 26, induced antibodies capable of neutralizing recent RSV A and RSV B clinical isolates, as well as protective efficacy against a challenge with RSV A and RSV B strains. Similarly, induction of cross-neutralizing antibodies was observed after immunization with Ad26-encoded preF, preF protein or a mix of both (Ad26/preF protein) in RSV pre-exposed mice and African Green Monkeys. Transfer of serum of human subjects immunized with Ad26/preF protein into cotton rats provide protection against challenges with both RSV A and RSV B, with complete protection against both strains observed in the lower respiratory tract. In contrast, almost no protection against RSV A and B infection was observed after the transfer of a human serum pool isolated pre-vaccination. These results collectively show that the RSV A-based monovalent Ad26/preF protein vaccine induced neutralizing antibodies, as well as protection against both RSV A and RSV B subtypes in animals, including by passive transfer of human antibodies alone, suggesting that clinical efficacy against both subtypes can be achieved.

2.
NPJ Vaccines ; 8(1): 45, 2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-36949051

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of severe respiratory disease for which no licensed vaccine is available. We have previously shown that a prefusion (preF) conformation-stabilized RSV F protein antigen and an adenoviral vector encoding RSV preF protein (Ad26.RSV.preF) are immunogenic and protective in animals when administered as single components. Here, we evaluated a combination of the 2 components, administered as a single injection. Strong induction of both humoral and cellular responses was shown in RSV-naïve and pre-exposed mice and pre-exposed African green monkeys (AGMs). Both components of the combination vaccine contributed to humoral immune responses, while the Ad26.RSV.preF component was the main contributor to cellular immune responses in both mice and AGMs. Immunization with the combination elicited superior protection against RSV A2 challenge in cotton rats. These results demonstrate the advantage of a combination vaccine and support further clinical development.

3.
Vaccine ; 40(6): 934-944, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-34973849

RESUMO

Respiratory Syncytial Virus (RSV) remains a leading cause of severe respiratory disease for which no licensed vaccine is available. We have previously described the derivation of an RSV Fusion protein (F) stabilized in its prefusion conformation (preF) as vaccine immunogen and demonstrated superior immunogenicity in naive mice of preF versus wild type RSV F protein, both as protein and when expressed from an Ad26 vaccine vector. Here we address the question if there are qualitative differences between the two vaccine platforms for induction of protective immunity. In naïve mice, both Ad26.RSV.preF and preF protein induced humoral responses, whereas cellular responses were only elicited by Ad26.RSV.preF. In RSV pre-exposed mice, a single dose of either vaccine induced cellular responses and strong humoral responses. Ad26-induced RSV-specific cellular immune responses were detected systemically and locally in the lungs. Both vaccines showed protective efficacy in the cotton rat model, but Ad26.RSV.preF conferred protection at lower virus neutralizing titers in comparison to RSV preF protein. Factors that may contribute to the protective capacity of Ad26.RSV.preF elicited immunity are the induced IgG2a antibodies that are able to engage Fcγ receptors mediating Antibody Dependent Cellular Cytotoxicity (ADCC), and the induction of systemic and lung resident RSV specific CD8 + T cells. These data demonstrate qualitative improvement of immune responses elicited by an adenoviral vector based vaccine encoding the RSV preF antigen compared to the subunit vaccine in small animal models which may inform RSV vaccine development.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Camundongos , Vírus Sincicial Respiratório Humano/genética , Proteínas Virais de Fusão/genética
4.
NPJ Vaccines ; 5(1): 49, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32566260

RESUMO

While RSV is a major cause of respiratory morbidity in infants, vaccine development is hindered by the immaturity and Th2-bias of the infant immune system and the legacy of enhanced respiratory disease (ERD) after RSV infection following immunization with formalin inactivated (FI)-RSV vaccine in earlier clinical trials. Preclinical studies have demonstrated that an adenoviral vector-based RSV F vaccine candidate (Ad26.RSV.FA2) induces Th1-biased protective immune responses, without signs of ERD upon subsequent RSV challenge. We here developed an Ad26 vector encoding the RSV F protein stabilized in its prefusion conformation (Ad26.RSV.preF). In adult mice, Ad26.RSV.preF induced superior, Th1-biased IgG2a-dominated humoral responses as compared to Ad26.RSV.FA2, while maintaining the strong Th1-biased cellular responses. Similar to adult mice, Ad26.RSV.preF induced robust and durable humoral immunity in neonatal mice, again characterized by IgG2a-dominated RSV F-binding antibodies, and high and stable virus-neutralizing titers. In addition, vaccine-elicited cellular immune responses were durable and characterized by IFN-γ-producing CD4+ and CD8+ T cells, with a profound Th1 bias. In contrast, immunization of neonatal mice with FI-RSV resulted in IgG1 RSV F-binding antibodies associated with a Th2 phenotype, no detectable virus-neutralizing antibodies, and a Th2-biased cellular response. These results are supportive for the clinical development of Ad26.RSV.preF for use in infants.

5.
PLoS One ; 13(8): e0202820, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30142207

RESUMO

In 2015, there was a large outbreak of Zika virus (ZIKV) in Brazil. Despite its relatively mild impact on healthy adults, ZIKV infection during pregnancy has been associated with severe birth defects. Currently, there is no ZIKV vaccine available, but several vaccine candidates based on the ZIKV membrane (M) and envelope (Env) structural proteins showed promising results in preclinical and clinical studies. Here, the immunogenicity and protective efficacy of a non-replicating adenoviral vector type 26 (Ad26) that encodes the ZIKV M-Env antigens (Ad26.ZIKV.M-Env) was evaluated in mice and non-human primates (NHP). Ad26.ZIKV.M-Env induced strong and durable cellular and humoral immune responses in preclinical models. Humoral responses were characterized by Env-binding and ZIKV neutralizing antibody responses while cellular responses were characterized by ZIKV reactive CD4+ and CD8+ T cells. Importantly, a single immunization with a very low dose of 4x107 vp of Ad26.ZIKV.M-Env protected mice from ZIKV challenge. In NHP, a single immunization with a typical human dose of 1x1011 vp of Ad26.ZIKV.M-Env also induced Env-binding and ZIKV neutralizing antibodies and Env and M specific cellular immune responses that associated with complete protection against viremia from ZIKV challenge as measured in plasma and other body fluids. Together these data provide the rationale to progress the Ad26.ZIKV.M-Env candidate vaccine to clinical testing.


Assuntos
Adenoviridae/genética , Antígenos Virais/genética , Imunidade Celular , Imunidade Humoral , Zika virus/patogenicidade , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vetores Genéticos/metabolismo , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Primatas , Vacinação , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Proteínas da Matriz Viral/metabolismo , Zika virus/metabolismo , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/veterinária , Infecção por Zika virus/virologia
6.
Int J Cancer ; 131(1): 117-28, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21823122

RESUMO

Altered glycosylation in epithelial cancers may play an important role in tumour progression, as it may affect tumour cell migration and antigen presentation by antigen presenting cells. We specifically characterise the glycosylation patterns of two tumour antigens that are highly expressed in cancer tissue and often detected in their secreted form in serum: the epithelial mucin MUC1 and carcinoembryonic antigen (CEA, also called CEACAM5). We analysed 48 colorectal cancer patients, comparing normal colon and tumour epithelium within each patient. Lectin binding was studied by a standardised CEA/MUC1 capture ELISA, using several plant lectins, and the human C-type lectins MGL and DC-SIGN, and Galectin-3. Peanut agglutinin (PNA) bound to MUC1 from tumour tissue in particular, suggests increased expression of the Thomsen-Friedenreich antigen (TF-antigen) (Core 1, Galß1-3GalNAc-Ser/Thr). Only small amounts of Tn-antigen (GalNAcα-Ser/Thr) expression was observed, but the human C-type lectin MGL showed increased binding to tumour-associated MUC1. Furthermore, sialylation was greatly enhanced. In sharp contrast, tumour-associated CEA (CEACAM5) contained high levels of the blood-group related carbohydrates, Lewis X and Lewis Y. This correlated strongly with the interaction of the human C-type lectin DC-SIGN to tumour-associated CEA, suggesting that CEA can be recognized and taken up by antigen presenting cells. In addition, increased mannose expression was observed and branched N-glycans were prominent, and this correlated well with human Galectin-3 binding. These data demonstrate that individual tumour antigens contain distinct glycan structures associated with cancer and, since glycans affect cellular interactions with its microenvironment, this may have consequences for progression of the disease.


Assuntos
Antígeno Carcinoembrionário/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Intestinal/metabolismo , Mucina-1/metabolismo , Antígenos Glicosídicos Associados a Tumores/biossíntese , Antígenos Glicosídicos Associados a Tumores/metabolismo , Moléculas de Adesão Celular/metabolismo , Neoplasias do Colo/patologia , Proteínas Ligadas por GPI/metabolismo , Galectina 3/metabolismo , Glicosilação , Humanos , Lectinas Tipo C/metabolismo , Antígenos do Grupo Sanguíneo de Lewis/biossíntese , Antígenos CD15/biossíntese , Manose/biossíntese , Aglutinina de Amendoim/metabolismo , Receptores de Superfície Celular/metabolismo
7.
Eur J Immunol ; 41(4): 916-25, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21400496

RESUMO

Cross-presentation is an important mechanism by which DCs present exogenous antigens on MHC-I molecules, and activate CD8(+) T cells, cells that are crucial for the elimination of tumors. We investigated the feasibility of exploiting the capacity of the mannose receptor (MR) to improve both cross-presentation of tumor antigens and Th polarization, processes that are pivotal for the anti-tumor potency of cytotoxic T cells. To this end, we selected two glycan ligands of the MR, 3-sulfo-Lewis(A) and tri-GlcNAc (N-acetylglucosamine), to conjugate to the model antigen OVA and assessed in vitro the effect on antigen presentation and Th differentiation. Our results demonstrate that conjugation of either 3-sulfo-Lewis(A) or tri-GlcNAc specifically directs antigen to the MR. Both neo-glycoconjugates showed, even at low doses, improved uptake as compared with native OVA, resulting in enhanced cross-presentation. Using MR(-/-) and MyD88-TRIFF(-/-) bone marrow-derived DCs (BMDCs), we show that the cross-presentation of the neo-glycoconjugates is dependent on MR and independent of TLR-mediated signaling. Whereas proliferation of antigen-specific CD4(+) T cells was unchanged, stimulation with neo-glycoconjugate-loaded DCs enhanced the generation of IFN-γ-producing T cells. We conclude that modification of antigen with either 3-sulfo-Lewis(A) or tri-GlcNAc enhances cross-presentation and permits Th1 skewing, through specific targeting of the MR, which may be beneficial for DC-based vaccination strategies to treat cancer.


Assuntos
Polaridade Celular , Apresentação Cruzada , Glicoconjugados/imunologia , Lectinas Tipo C/imunologia , Lectinas de Ligação a Manose/imunologia , Oligossacarídeos/imunologia , Receptores de Superfície Celular/imunologia , Células Th1/imunologia , Trissacarídeos/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Células Cultivadas , Endossomos/imunologia , Lectinas Tipo C/deficiência , Antígenos do Grupo Sanguíneo de Lewis , Receptor de Manose , Lectinas de Ligação a Manose/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/deficiência , Células Th1/citologia , Receptores Toll-Like/imunologia
8.
Biochem Soc Trans ; 39(1): 388-92, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21265810

RESUMO

A common phenotypic change in cancer is a dramatic transformation of cellular glycosylation. Functional studies of particular tumour-associated oligosaccharides are difficult to interpret conclusively, but carbohydrate-binding proteins are likely to contribute to progression of the tumour. This review discusses the potential role of CLRs (C-type lectin receptors), expressed by antigen-presenting cells of the immune system, in tumour recognition and immune modulation. Studies in recent years have provided significant insight into the immunomodulatory function of CLR during infections, but their role in cancer remains elusive; some strongly bind tumour cells and antigens, indicating participation in malignancy. The potential to use recombinant CLR as diagnostic tools will also be discussed.


Assuntos
Antígenos de Neoplasias/imunologia , Sistema Imunitário/imunologia , Animais , Antígenos de Neoplasias/química , Biomarcadores Tumorais/química , Biomarcadores Tumorais/imunologia , Células Dendríticas/imunologia , Glicoproteínas/química , Glicoproteínas/imunologia , Glicosilação , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Oligossacarídeos/química , Oligossacarídeos/imunologia , Polissacarídeos/química , Polissacarídeos/imunologia
9.
Int J Cancer ; 128(6): 1371-83, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20473945

RESUMO

We recently showed that MGL2 specifically binds tumour-associated glycan N-acetylgalactosamine (GalNAc). We here demonstrate that modification of an antigen with tumour-associated glycan GalNAc, targets antigen specifically to the MGL2 on bone marrow derived (BM)-DCs and splenic DCs. Glycan-modification of antigen with GalNAc that mimics tumour-associated glycosylation, promoted antigen internalisation in DCs and presentation to CD4 T cells, as well as differentiation of IFN-γ producing CD4 T cells. Furthermore, GalNAc modified antigen enhanced cross-presentation of both BM-DCs and primary splenic DCs resulting in enhanced antigen specific CD8 T cell responses. Using MyD88-TRIFF(-/-) BM-DCs we demonstrate that the enhanced cross-presentation of the GalNAc modified antigen is TLR independent. Our data strongly suggest that tumour-associated GalNAc modification of antigen targets MGL on DCs and greatly enhances both MHC class II and class I presentation in a TLR independent manner.


Assuntos
Acetilgalactosamina/metabolismo , Antígenos Glicosídicos Associados a Tumores/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Lectinas Tipo C/fisiologia , Animais , Apresentação de Antígeno/imunologia , Western Blotting , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células CHO , Proliferação de Células , Cricetinae , Cricetulus , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Glicosilação , Antígenos de Histocompatibilidade Classe I/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/fisiologia , Ovalbumina/fisiologia , RNA Mensageiro/genética , Receptores de Antígenos de Linfócitos T/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Baço/metabolismo
10.
Mol Immunol ; 47(2-3): 164-74, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19818504

RESUMO

Dendritic cells have gained much interest in the field of anti-cancer vaccine development because of their central function in immune regulation. One of the receptors that facilitate DC-specific targeting of antigens is the DC-specific C-type lectin DC-SIGN. Although DC-SIGN is specifically expressed on human DCs, its murine homologue is not present on any murine DC subsets, which makes in vivo evaluation of potential DC-SIGN targeting vaccines very difficult. Here we describe the use of DC-SIGN transgenic mice, as a good model system to evaluate DC-SIGN targeting vaccines. We demonstrate that glycan modification of OVA with DC-SIGN targeting glycans, targets antigen specifically to bone marrow (BM)** derived DCs and splenic DCs. Glycan modification of OVA with Lewis X or Lewis B oligosaccharides, that target DC-SIGN transgenic DCs, resulted in efficient 10-fold induction of OT-II compared to unmodified OVA. Interestingly, glycan modified OVA proteins were significantly cross-presented to OT-I T cells by wild type DC, 10-fold more than native OVA, and the expression of DC-SIGN further enhanced this cross-presentation. Targeting of glycosylated OVA was neither accompanied with any DC maturation, nor the production of inflammatory or anti-inflammatory cytokines. Thus, we conclude that glycan modification of antigens and targeting to DC-SIGN enhance both CD4 and CD8 T cell responses. Furthermore, our data demonstrate that DC-SIGN transgenic mice are valuable tool for optimisation and efficiency testing of DC vaccination strategies that are designed to target in particular the human DC-SIGN receptor.


Assuntos
Apresentação de Antígeno/imunologia , Moléculas de Adesão Celular/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Lectinas Tipo C/imunologia , Ovalbumina/imunologia , Polissacarídeos/imunologia , Receptores de Superfície Celular/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Metabolismo dos Carboidratos/imunologia , Apresentação Cruzada/imunologia , Glicoconjugados/imunologia , Humanos , Antígenos CD15/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
11.
Cell Microbiol ; 11(12): 1768-81, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19681908

RESUMO

An increasing number of bacterial pathogens produce an array of glycoproteins of unknown function. Here we report that Campylobacter jejuni proteins that are modified by the N-linked glycosylation machinery encoded by the pgl locus bind the human Macrophage Galactose-type lectin (MGL). MGL receptor binding was abrogated by EDTA and N-acetylgalactosamine (GalNAc) and was successfully transferred to Escherichia coli by introducing the C. jejuni pgl locus together with a glycan acceptor protein. In addition to glycoproteins, C. jejuni lipooligosaccharide with a terminal GalNAc residue was recognized by MGL. Recombinant E. coli expressing the C. jejuni pgl locus in the absence of a suitable glycan acceptor protein produced altered lipopolysaccharide glycoforms that gained MGL reactivity. Infection assays demonstrated high levels of GalNAc-dependent interaction of the recombinant E. coli with MGL-transfected mammalian cells. In addition, interleukin-6 production by human dendritic cells was enhanced by C. jejuni lacking N-linked glycans compared with wild-type bacteria. Collectively, our results provide evidence that both N-linked glycoproteins and distinct lipooligosaccharide glycoforms of C. jejuni are ligands for the human C-type lectin MGL and that the C. jejuni N-glycosylation machinery can be exploited to target recombinant bacteria to MGL-expressing eukaryotic cells.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Campylobacter/metabolismo , Campylobacter jejuni/metabolismo , Glicoproteínas/metabolismo , Lectinas Tipo C/metabolismo , Lipopolissacarídeos/metabolismo , Acetilgalactosamina/metabolismo , Acetilgalactosamina/farmacologia , Animais , Proteínas de Bactérias/genética , Células CHO , Infecções por Campylobacter/microbiologia , Campylobacter jejuni/efeitos dos fármacos , Campylobacter jejuni/genética , Quelantes/farmacologia , Cricetinae , Cricetulus , Células Dendríticas/metabolismo , Ácido Edético/farmacologia , Escherichia coli/metabolismo , Glicosilação , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Interleucina-6/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
12.
Mol Immunol ; 46(6): 1240-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19162326

RESUMO

Antigen presenting cells (APC) express a variety of pattern recognition receptors, including the C-type lectin receptors (CLR) that specifically recognize carbohydrate structures expressed on self-tissue and pathogens. The CLR play an important role in antigen uptake and presentation and have been shown to mediate cellular interactions. The ligand specificity of the human macrophage galactose-type lectin (MGL) has been characterized extensively. Here, we set out to determine the glycan specificity of the murine homologues, MGL1 and MGL2, using a glycan array. Murine MGL1 was found to be highly specific for Lewis X and Lewis A structures, whereas mMGL2, more similar to the human MGL, recognized N-acetylgalactosamine (GalNAc) and galactose, including the O-linked Tn-antigen, TF-antigen and core 2. The generation of MGL1 and MGL2-Fc proteins allowed us to identify ligands in lymph nodes, and MGL1-Fc additionally recognized high endothelial venules. Strikingly, MGL2 interacted strongly to adenocarcinoma cells, suggesting a potential role in tumor immunity.


Assuntos
Assialoglicoproteínas/metabolismo , Lectinas Tipo C/metabolismo , Proteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Adenocarcinoma/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Assialoglicoproteínas/genética , Assialoglicoproteínas/imunologia , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Ligantes , Linfonodos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pele/metabolismo
13.
Trends Immunol ; 29(2): 83-90, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18249034

RESUMO

C-type lectins play important roles in both innate and adaptive immune responses. In contrast to the mannose- or fucose-specific C-type lectins DC-SIGN and mannose receptor, the galactose-type lectins, of which only macrophage galactose-type lectin (MGL) is found within the immune system, are less well known. MGL is selectively expressed by immature dendritic cells and macrophages with elevated levels on tolerogenic or alternatively activated subsets. Human MGL has an exclusive specificity for rare terminal GalNAc structures, which are revealed on the tumor-associated mucin MUC1 and CD45 on effector T cells. These findings implicate MGL in the homeostatic control of adaptive immunity. We discuss here the functional similarities and differences between MGL orthologs and compare MGL to its closest homolog, the liver-specific asialoglycoprotein receptor (ASGP-R).


Assuntos
Carboidratos/imunologia , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Animais , Apresentação de Antígeno , Receptor de Asialoglicoproteína/imunologia , Receptor de Asialoglicoproteína/metabolismo , Carboidratos/química , Células Dendríticas/imunologia , Humanos , Lectinas Tipo C/imunologia , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/imunologia , Neoplasias/imunologia
14.
Cancer Immunol Immunother ; 56(8): 1225-36, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17195076

RESUMO

The epithelial mucin MUC1 is a high molecular weight membrane glycoprotein frequently overexpressed and aberrantly glycosylated in adenocarcinoma. Mucins normally contain high amounts of O-linked carbohydrate structures that may influence immune reactions to this antigen. During malignant transformation, certain glyco-epitopes of MUC1, such as Tn-antigen, TF-antigen and their sialylated forms become exposed. The role of these glycan structures in tumor biology is unknown, but their presence is known to correlate with poor prognosis in several adenocarcinomas. We analyzed the potency of MUC1 containing Tn-antigens (MUC1-Tn) to target C-type lectins that function as carbohydrate recognition and uptake molecules on dendritic cells (DC). We identified the macrophage galactose type C-type lectin (MGL), expressed by both DC and macrophages, as the receptor for recognition and binding of MUC1-Tn. To validate the occurrence of MGL-MUC1 interactions in situ, we studied the binding of MGL to MUC1 in primary colon carcinoma tissue. Isolation of MUC1 out of colon carcinoma tissue showed strong binding activity to MGL. Interestingly, MGL binding to MUC1 was highly correlated to binding by the lectin Helix pomatia agglutinin (HPA), which is associated with poor prognosis in colorectal cancer. The detection of MGL positive cells in situ at the tumor site together with the modified glycosylation status of MUC1 to target MGL on DC suggests that MGL positive antigen presenting cells may play a role in tumor progression.


Assuntos
Adenocarcinoma/metabolismo , Antígenos de Neoplasias/metabolismo , Antígenos Glicosídicos Associados a Tumores/metabolismo , Neoplasias do Colo/metabolismo , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Mucinas/metabolismo , Proteínas de Neoplasias/metabolismo , Acetilgalactosamina/farmacologia , Adenocarcinoma/imunologia , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/isolamento & purificação , Antígenos Glicosídicos Associados a Tumores/imunologia , Células CHO , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Colo/química , Colo/citologia , Neoplasias do Colo/imunologia , Cricetinae , Cricetulus , Células Dendríticas/imunologia , Endocitose , Feminino , Glicosilação , Humanos , Imunoglobulina G/imunologia , Mucosa Intestinal/química , Mucosa Intestinal/citologia , Lectinas Tipo C/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/citologia , Monossacarídeos/farmacologia , Mucina-1 , Mucinas/imunologia , Mucinas/isolamento & purificação , Proteínas de Neoplasias/imunologia , Proteínas de Neoplasias/isolamento & purificação , Ligação Proteica/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Sequências de Repetição em Tandem
15.
Curr Opin Immunol ; 18(1): 105-11, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16303292

RESUMO

C-type lectin receptors on antigen-presenting cells are potent antigen-uptake receptors with specificity for glycan structures. Glycosylation changes during malignant transformation create tumor-specific carbohydrate structures that interact with C-type lectins on dendritic cells. Recent findings revealed that tumor glycoproteins, such as carcinoembryonic antigen and MUC-1, indeed interact with the C-type lectins DC-SIGN and macrophage galactose-type lectin on antigen-presenting cells. The consequences for anti-cancer immunity or tolerance induction can be extrapolated from the function of C-type lectins in pathogen recognition and antigen presentation. In addition, in vivo studies in mice recently demonstrated the potency of targeting antigens to C-type lectins on antigen-presenting cells for anti-tumor vaccination strategies.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Antígenos de Neoplasias/imunologia , Neoplasias/imunologia , Polissacarídeos/imunologia , Receptores de Superfície Celular/imunologia , Animais , Células Apresentadoras de Antígenos/química , Antígeno Carcinoembrionário/imunologia , Glicosilação , Humanos , Imunoterapia , Lectinas Tipo C/química , Camundongos , Neoplasias/prevenção & controle , Neoplasias/terapia
16.
Int Immunol ; 17(5): 661-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15802303

RESUMO

Dendritic cells (DCs) are key to the maintenance of peripheral tolerance to self-antigens and the orchestration of an immune reaction to foreign antigens. C-type lectins, expressed by DCs, recognize carbohydrate moieties on antigens that can be internalized for processing and presentation. Little is known about the exact glycan structures on self-antigens and pathogens that are specifically recognized by the different C-type lectins and how this interaction influences DC function. We have analyzed the carbohydrate specificity of the human C-type lectin macrophage galactose-type lectin (MGL) using glycan microarray profiling and identified an exclusive specificity for terminal alpha- and beta-linked GalNAc residues that naturally occur as parts of glycoproteins or glycosphingolipids. Specific glycan structures containing terminal GalNAc moieties, expressed by the human helminth parasite Schistosoma mansoni as well as tumor antigens and a subset of gangliosides, were identified as ligands for MGL. Our results indicate an endogenous function for DC-expressed MGL in the clearance and tolerance to self-gangliosides, and in the pattern recognition of tumor antigens and foreign glycoproteins derived from helminth parasites.


Assuntos
Antígenos de Helmintos/imunologia , Antígenos Glicosídicos Associados a Tumores/imunologia , Carboidratos/imunologia , Células Dendríticas/imunologia , Lectinas Tipo C/metabolismo , Schistosoma mansoni/imunologia , Animais , Apresentação de Antígeno , Linhagem Celular Tumoral , Células Cultivadas , Galactose/imunologia , Glucose/imunologia , Glicoproteínas/imunologia , Glicoesfingolipídeos/imunologia , Glicoesfingolipídeos/metabolismo , Humanos , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA