Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Crit Rev Immunol ; 37(2-6): 421-437, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29773028

RESUMO

Mucins are attracting great interest as potential targets for immunotherapy in the development of vaccines for cancers expressing Mucinl (MUC1) (e.g., breast, pancreas, ovary, and others) as there is (1) a 10-fold increase in the amount in adenocarcinomas; (2) an alteration in expression where they become ubiquitous, and (3) due to altered glycosylation, new epitopes appear on the cell surface that are absent in normal tissues. These new epitopes can be carbohydrate; others are peptide in nature. The cloning of the cDNAs from mucins, particularly MUC1, has led to rapid advances being made, and it is clear that a highly immunogenic peptide exists within the variable number of tandem repeats (VNTR) found in all mucins. This peptide is immunogenic in mice, giving rise to strong antibody production, and most monoclonal antibodies made to breast cancer, which react with the protein core, react with the peptide APDTR. It is now also clear that humans with breast cancer have, in their draining lymph nodes, precursors of cytotoxic T cells that can be stimulated in vitro to react against breast cancer and indeed against the APDTR or a closely related peptide - shown from antibody-blocking studies. These CTLs are unique in that they are non-MHC restricted. The identification of suitable targets, coupled with the known immunogenicity of both the peptide and neo-carbohydrate epitopes, has led to the development of several different programs to immunize humans against breast cancer using either synthetic carbohydrates or peptides conjugated with adjuvants, and clinical trials are now in progress to evaluate their immunogenicity and anti-cancer effects.


Assuntos
Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Imunoterapia/métodos , Mucinas/imunologia , Neoplasias/terapia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Vacinas Anticâncer/genética , Ensaios Clínicos como Assunto , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitopos/genética , Epitopos/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Glicosilação , Humanos , Imunogenicidade da Vacina/genética , Imunogenicidade da Vacina/imunologia , Camundongos , Repetições Minissatélites/imunologia , Mucinas/genética , Mucinas/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Vacinação/métodos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-27442413

RESUMO

Immunotherapy using mucin 1 (MUC1) linked to oxidised mannan (MFP) was investigated in an aggressive MUC1+ metastatic tumour, DA3-MUC1 because, unlike many MUC1+ tumour models, DA3-MUC1 is not spontaneously rejected in mice making it an alternative model for immunotherapy studies. Further, DA3-MUC1 cells are resistant to lysis by anti-MUC1 cytotoxic T cells (CTLs). The inability of DA3-MUC1 tumours to be rejected in naïve mice as well as vaccination to MUC1 was attributed to a deficiency of expression of MHC class I molecules on the tumour cell surface. In vitro and in vivo analysis of subcutaneous tumours and lung metastases demonstrated that DA3-MUC1 tumour cells have a low expression (< 6%) of MHC class I which can be upregulated (> 90%) following culturing with IFN-γ. Results from flow cytometry analysis and immunoperoxidase staining indicated that the in vitro up-regulation of MHC class I could be maintained for up to seven days in vivo, without affecting the expression levels of MUC1 antigen. Interestingly, MUC1-specific CTL that lyse DA3-MUC1 targets in vitro were induced in MFP immunised mice but failed to protect mice from a DA3-MUC1 tumour challenge. These results highlight the importance of MHC class I molecules in the induction of anti-tumour immunity and the MFP immune response.


Assuntos
Adenocarcinoma/etiologia , Adenocarcinoma/secundário , Neoplasias da Mama/patologia , Imunoterapia , Interferon gama/fisiologia , Mucina-1/imunologia , Animais , Neoplasias da Mama/etiologia , Feminino , Antígenos de Histocompatibilidade Classe I , Camundongos , Camundongos Endogâmicos C57BL
4.
Immunotherapy ; 5(11): 1177-82, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24188672

RESUMO

BACKGROUND: Targeting antigens to dendritic cell receptors has recently become a popular approach to inducing effective immune responses against cancer antigens. Almost 20 years ago, however, we demonstrated that targeting the mannose receptor on macrophages and dendritic cells leads to strong cellular immune responses. We conducted numerous human clinical trials demonstrating the effectiveness of oxidized mannan-MUC1 (M-FP) in MUC1(+) adenocarcinoma patients. In one trial, the 5-8-year follow-up of breast cancer patients vaccinated with M-FP was published previously; we now report here the 12-15-year follow-up. Details regarding the preparation of the vaccine, inclusion and exclusion criteria, immunotherapy and follow-up schedule, were published previously. RESULTS: The follow-up at 12-15 years showed that the recurrence rate in patients receiving placebo was 60% (nine of 15). In those receiving immunotherapy (M-FP), the rate was 12.5% (two of 16). The time of recurrence in the placebo group ranged from 7 to 180 months (mean: 65.8 months) and in the two patients of the vaccine group, the recurrence appeared at 95 and 141 months (mean: 118 months) after surgery. These findings are statistically significant (p = 0.02 for survival and p = 0.009 for percentage of patients cancer-free). All patients injected with M-FP showed no evidence of toxic effects or signs of autoimmunity during the 12-15-year follow-up. DISCUSSION: The preliminary evidence indicates that M-FP is beneficial in the overall survival of early-stage breast cancer patients. This long-term clinical follow-up of patients strongly supports the necessity for a large Phase III study of direct M-FP injection in early-stage breast cancer patients, to evaluate immunotherapy as an adjuvant treatment for breast cancer.


Assuntos
Adenocarcinoma , Neoplasias da Mama , Vacinas Anticâncer , Imunoterapia , Mananas , Mucina-1 , Adenocarcinoma/imunologia , Adenocarcinoma/mortalidade , Adenocarcinoma/terapia , Idoso , Neoplasias da Mama/imunologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/terapia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Mananas/administração & dosagem , Mananas/imunologia , Pessoa de Meia-Idade , Mucina-1/administração & dosagem , Mucina-1/imunologia , Oxirredução , Taxa de Sobrevida
6.
Immunol Cell Biol ; 85(8): 623-32, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17724458

RESUMO

Carbohydrates are involved in many immunological responses including the rejection of incompatible blood, tissues and organs. Carbohydrate antigens with Galalpha(1,3)Gal epitopes are recognized by natural antibodies in humans and pose a major barrier for pig-to-human xenotransplantation. Genetically modified pigs have been established that have no functional alpha1,3-galactosyltransferase (alpha1,3GT), which transfers alphaGal to N-acetyllactosamine (LacNAc) type oligosaccharides. However, a low level of Galalpha(1,3)Gal is still expressed in alpha1,3GT knockout animals in the form of a lipid, isoglobotrihexosylceramide (iGb3), which is produced by iGb3 synthase on lactose (Lac) type core structures. Here, we define the reactivity of a series of monoclonal antibodies (mAb) generated in alpha1,3GT-/- mice immunized with rabbit red blood cells (RbRBC), as a rich source of lipid-linked antigens. Interestingly, one mAb (15.101) binds weakly to synthetic and cell surface-expressed Galalpha(1,3)Gal on LacNAc, but strongly to versions of the antigen on Lac cores, including iGb3. Three-dimensional models suggest that the terminal alpha-linked Gal binds tightly into the antibody-binding cavity. Furthermore, antibody interactions were predicted with the second and third monosaccharide units. Collectively, our findings suggest that although the terminal carbohydrate residues confer most of the binding affinity, the fine specificity is determined by subsequent residues in the oligosaccharide.


Assuntos
Anticorpos Heterófilos/imunologia , Especificidade de Anticorpos/imunologia , Carboidratos/imunologia , Dissacarídeos/imunologia , Epitopos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Antígenos/imunologia , Sítios de Ligação de Anticorpos , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Glicoconjugados/imunologia , Humanos , Ligação de Hidrogênio , Camundongos , Modelos Moleculares , Oligossacarídeos/imunologia
7.
Immunology ; 120(3): 325-35, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17328786

RESUMO

Recent years have seen a resurgence in interest in the development of efficient non-viral delivery systems for DNA vaccines and gene therapy. We have previously used oxidized and reduced mannan as carriers for protein delivery to antigen-presenting cells by targeting the receptors that bind mannose, resulting in efficient induction of cellular responses. In the present study, oxidized mannan and reduced mannan were used as receptor-mediated gene transfer ligands for cancer immunotherapy. In vivo studies in C57BL/6 mice showed that injection of DNA encoding ovalbumin (OVA) complexed to oxidized or reduced mannan-poly-L-lysine induced CD8 and CD4 T-cell responses as well as antibody responses leading to protection of mice from OVA+ tumours. Both oxidized and reduced mannan delivery was superior to DNA alone or DNA-poly-L-lysine. These studies demonstrate the potential of oxidized and reduced mannan for efficient receptor-mediated gene delivery in vivo, particularly as DNA vaccines for cancer immunotherapy.


Assuntos
Vacinas Anticâncer/imunologia , Técnicas de Transferência de Genes , Mananas/imunologia , Neoplasias Experimentais/prevenção & controle , Vacinas de DNA/imunologia , Animais , Anticorpos Antineoplásicos/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Terapia Genética/métodos , Lectinas Tipo C/imunologia , Ativação Linfocitária/imunologia , Receptor de Manose , Lectinas de Ligação a Manose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Experimentais/imunologia , Ovalbumina/genética , Ovalbumina/imunologia , Oxirredução , Polilisina/imunologia , Polilisina/farmacologia , Receptores de Superfície Celular/imunologia
8.
Vaccine ; 24(16): 3191-202, 2006 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-16480791

RESUMO

Cytoplasmic delivery of proteins or CTL epitopes is crucial for the presentation of antigen for the generation of CTL. We previously described the use of the 16-amino acid peptide penetratin from the Drosophila Antennapedia domain (Int) to transport CTL epitopes into cells. Here we show that, Int, incorporating MUC1 CTL epitopes in tandem is able to facilitate their rapid uptake by macrophages and dendritic cells (DC) in an energy-dependent endocytic pathway. We also demonstrate for the first time that Int conjugated proteins are also able to be efficiently taken up by DC. Furthermore, C57BL/6 and HLA-A2 transgenic mice immunized with the Int-peptides or Int-proteins induce strong IFN-gamma secreting T cells and weak IgG1 antibodies. Immunized C57BL/6 mice were protected against the growth of a MUC1(+) tumor cell line.


Assuntos
Apresentação de Antígeno , Vacinas Anticâncer/imunologia , Proteínas de Transporte/farmacologia , Mucina-1/imunologia , Neoplasias Experimentais/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Antineoplásicos/sangue , Proteínas de Transporte/administração & dosagem , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Peptídeos Penetradores de Células , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Antígeno HLA-A2/genética , Imunoglobulina G/sangue , Interferon gama/biossíntese , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Mucina-1/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo
9.
Clin Cancer Res ; 12(3 Pt 1): 869-77, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16467101

RESUMO

PURPOSE: Tumor antigen-loaded dendritic cells show promise for cancer immunotherapy. This phase I study evaluated immunization with autologous dendritic cells pulsed with mannan-MUC1 fusion protein (MFP) to treat patients with advanced malignancy. EXPERIMENTAL DESIGN: Eligible patients had adenocarcinoma expressing MUC1, were of performance status 0 to 1, with no autoimmune disease. Patients underwent leukapheresis to generate dendritic cells by culture ex vivo with granulocyte macrophage colony-stimulating factor and interleukin 4 for 5 days. Dendritic cells were then pulsed overnight with MFP and harvested for reinjection. Patients underwent three cycles of leukapheresis and reinjection at monthly intervals. Patients with clinical benefit were able to continue with dendritic cell-MFP immunotherapy. RESULTS: Ten patients with a range of tumor types were enrolled, with median age of 60 years (range, 33-70 years); eight patients were of performance status 0 and two of performance status 1. Dendritic cell-MFP therapy led to strong T-cell IFNgamma Elispot responses to the vaccine and delayed-type hypersensitivity responses at injection sites in nine patients who completed treatments. Immune responses were sustained at 1 year in monitored patients. Antibody responses were seen in three patients only and were of low titer. Side effects were grade 1 only. Two patients with clearly progressive disease (ovarian and renal carcinoma) at entry were stable after initial therapy and went on to further leukapheresis and dendritic cell-MFP immunotherapy. These two patients have now each completed over 3 years of treatment. CONCLUSIONS: Immunization produced T-cell responses in all patients with evidence of tumor stabilization in 2 of the 10 advanced cancer patients treated. These data support further clinical evaluation of this dendritic cell-MFP immunotherapy.


Assuntos
Adenocarcinoma/terapia , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Imunoterapia , Mananas/administração & dosagem , Mucinas/administração & dosagem , Adenocarcinoma/imunologia , Adulto , Idoso , Antígenos de Neoplasias , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/toxicidade , Células Dendríticas/transplante , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Esquemas de Imunização , Interferon gama/imunologia , Interferon gama/metabolismo , Leucaférese , Masculino , Mananas/imunologia , Mananas/toxicidade , Pessoa de Meia-Idade , Mucina-1 , Mucinas/imunologia , Fenótipo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T/imunologia , Resultado do Tratamento
10.
Mol Cancer Res ; 3(3): 170-81, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15798097

RESUMO

Elevated expression of the serine/threonine kinase Pim-1 increases the incidence of lymphomas in Pim-1 transgenic mice and has also been found to occur in some human cancers. Pim-1 acts as a cell survival factor and may prevent apoptosis in malignant cells. It was therefore of interest to understand to what extent maintenance and degradation of Pim-1 protein is affected by heat shock proteins (Hsp) and the ubiquitin-proteasome pathway in K562 and BV173 human leukemic cells. The half-life of Pim-1 protein in these cells was found to increase from 1.7 to 3.1 hours when induced by heat shock or by treating the cells with the proteasome inhibitor PS-341 (bortezomib). The Hsp90 inhibitor geldanamycin prevented the stabilization of Pim-1 by heat shock. Using immunoprecipitation, it was determined that Pim-1 is targeted for degradation by ubiquitin and that Hsp70 is associated with Pim-1 under these circumstances. Conversely, Hsp90 was found to protect Pim-1 from proteasomal degradation. A luminescence-based kinase assay showed that Pim-1 kinase bound to Hsp70 or Hsp90 remains active, emphasizing the importance of its overall cellular levels. This study shows how Pim-1 levels can be modulated in cells through degradation and stabilization.


Assuntos
Proteínas de Choque Térmico/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Ubiquitina/química , Trifosfato de Adenosina/química , Animais , Apoptose , Benzoquinonas , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Eletroporação , Citometria de Fluxo , Proteínas de Choque Térmico HSP70/química , Proteínas de Choque Térmico HSP90/química , Temperatura Alta , Humanos , Immunoblotting , Imunoprecipitação , Células K562 , Lactamas Macrocíclicas , Camundongos , Camundongos Transgênicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas c-pim-1 , Pirazinas/farmacologia , Quinonas/farmacologia , Fatores de Tempo , Ubiquitina/metabolismo
11.
J Immunol ; 173(5): 3148-54, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15322175

RESUMO

Infection can protect against subsequent disease by induction of both humoral and cellular immunity, but inert protein-based vaccines are not as effective. In this study, we present a new vaccine design, with Ag covalently conjugated to solid core nano-beads of narrowly defined size (0.04-0.05 microm) that localize to dendritic cells (DEC205(+) CD40(+), CD86(+)) in draining lymph nodes, inducing high levels of IFN-gamma production (CD8 T cells: precursor frequencies 1/5000 to 1/1000) and high Ab titers in mice. Conjugation of Ag to these nano-beads induced responses that were significantly higher (2- to 10-fold) than those elicited by other bead sizes, and higher than a range of currently used adjuvants (alum, QuilA, monophosphoryl lipid A). Responses were comparable to CFA/IFA immunization for Abs and ex vivo peptide-pulsed dendritic cell immunization for CD8 T cells. A single dose of Ag-conjugated beads protected mice from tumors in two different model challenges and caused rapid clearance of established tumors in mice. Thus, a range of Ags conjugated to nano-beads was effective as immunogens in both therapeutic and prophylactic scenarios.


Assuntos
Vacinas Anticâncer/imunologia , Nanotecnologia , Neoplasias/prevenção & controle , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos/imunologia , Vacinas Anticâncer/farmacologia , Modelos Animais de Doenças , Camundongos , Nanotubos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia
12.
Cancer Res ; 64(11): 4018-23, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15173016

RESUMO

Cripto, a member of the epidermal growth factor-Cripto-FRL-Criptic (EGF-CFC) family, has been described recently as a potential target for immunotherapy (Adkins et al., J Clin Invest 2003;112:575-87). We have produced rat monoclonal antibodies (mAbs) to a Cripto 17-mer peptide, corresponding to the "EGF-like" motif of Cripto. The mAbs react with most cancers of the breast, colon, lung, stomach, and pancreas but do not react or react weakly with normal tissues. The mAbs inhibit cancer cell growth in vitro, and this effect was greater with cytotoxic drugs such as 5-fluorouracil, epirubicin, and cisplatin. The anti-Cripto mAbs prevent tumor development in vivo and inhibit the growth of established tumors of LS174T colon xenografts in Scid mice. The growth inhibitory effects with these mAbs may be greater than those described elsewhere, possibly because of IgM giving more effective cross-linking or binding to a different epitope (EGF-like region versus CFC region). The mechanism of inhibitory effects of the Cripto mAbs includes both cancer cell apoptosis, activation of c-Jun-NH(2)-terminal kinase and p38 kinase signaling pathways and blocking of Akt phosphorylation. Thus, Cripto is a unique target, and mAbs to Cripto could be of therapeutic value for human cancers.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Fator de Crescimento Epidérmico/antagonistas & inibidores , Fator de Crescimento Epidérmico/imunologia , Imunização Passiva/métodos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Linhagem Celular Tumoral , Feminino , Proteínas Ligadas por GPI , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia , Ratos , Ratos Endogâmicos Lew , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Xenotransplantation ; 11(2): 171-83, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14962279

RESUMO

Human membrane cofactor protein (CD46) controls complement activation and when expressed sufficiently as a transgene protects xenografts against complement-mediated rejection, as shown here using non-immunosuppressed baboons and heterotopic CD46 transgenic pig kidney xenografts. This report is of a carefully engineered transgene that enables high-level CD46 expression. A novel CD46 minigene was validated by transfection and production of a transgenic pig line. Pig lymphocytes were tested for resistance to antibody and complement-mediated lysis, transgenic tissues were characterized for CD46 expression, and kidneys were transplanted to baboons without immunosuppression. Absorption of anti-Galalpha(1,3)Gal epitope (anti-GAL) serum antibodies was measured. Transgenic pigs expressed high levels of CD46 in all tissues, especially vascular endothelium, with stable expression through three generations that was readily monitored by flow cytometry of transgenic peripheral blood mononuclear cells (PBMC). Transgenic PBMC pre-sensitized with antibody were highly resistant to human complement-mediated lysis which readily lysed normal pig PBMC. Normal pig kidneys transplanted without cold ischemia into non-immunosuppressed adult baboons survived a median of 3.5 h (n = 7) whereas transgenic grafts (n = 9), harvested at approximately 24-h intervals, were either macroscopically normal (at 29, 48 and 68 h) or showed limited macroscopic damage (median > 50 h). Microscopic assessment of transplanted transgenic kidneys showed only focal tubular infarcts with viable renal tissue elsewhere, no endothelial swelling or polymorph adherence and infiltration by lymphocytes beginning at 3 days. Coagulopathy was not a feature of the histology in four kidneys not rejected and assessed at 48 h or later after transplantation. Baboon anti-GAL serum antibody titers were high before transplantation and, in one extensively analyzed recipient, reduced approximately 8-fold within 5.5 h. The data demonstrate that a single CD46 transgene controls hyperacute kidney graft rejection in untreated baboons despite the presence of antibody and complement deposition. The expression levels, tissue distribution and in vitro functional tests indicate highly efficient CD46 function, controlling both classical and alternative pathway complement activation, which suggests it might be the complement regulator of choice to protect xenografts.


Assuntos
Antígenos CD/genética , Rejeição de Enxerto/imunologia , Transplante de Rim/imunologia , Glicoproteínas de Membrana/genética , Transplante Heterólogo/imunologia , Doença Aguda , Animais , Animais Geneticamente Modificados , Anticorpos Heterófilos/sangue , Cruzamentos Genéticos , Dissacarídeos/sangue , Epitopos/sangue , Rejeição de Enxerto/prevenção & controle , Humanos , Terapia de Imunossupressão , Transplante de Rim/patologia , Proteína Cofatora de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Papio , Suínos , Transplante Heterólogo/patologia
15.
Proc Natl Acad Sci U S A ; 100(25): 15029-34, 2003 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-14657390

RESUMO

Peptides bind MHC class I molecules by anchoring hydrophobic side chains into pockets in the peptide binding groove. Here, we report an immunogenic (in vitro and in vivo) MUC1 glycopeptide (MUC1-8-5GalNAc) bound to H-2Kb, fully crossreactive with the nonglycosylated variant. Molecular modeling showed that the central P5-Thr-GalNAc residue points into the C pocket and forms van der Waals and hydrogen bond interactions with the MHC class I. As predicted, GalNAc, a modified peptide carrying an additional anchor in the central C anchor pocket, increased the affinity by approximately 100-fold compared with the native low-affinity peptide (MUC1-8). The findings demonstrate that glycopeptides associated with MHC class I molecules can use GalNAc to anchor the peptide in the groove and enable high-affinity binding.


Assuntos
Acetilgalactosamina/química , Genes MHC Classe I , Glicopeptídeos/química , Animais , Sítios de Ligação , Ensaio de Imunoadsorção Enzimática , Feminino , Ligação de Hidrogênio , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Peptídeos/química , Ligação Proteica , Estrutura Secundária de Proteína , Temperatura , Fatores de Tempo
16.
Glycobiology ; 13(5): 327-37, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12626403

RESUMO

The important xenoepitope Galalpha(1,3)Gal was thought to be exclusively synthesized by a single alpha(1,3)galactosyltransferase. However, the cloning of the distant family member rat iGb3 synthase, which is also capable of synthesizing Galalpha(1,3)Gal as the glycolipid structure iGb3, challenges the notion that alpha(1,3)galactosyltransferase is the sole Galalpha(1,3)Gal-synthesizing enzyme. We describe the cloning of the rat homolog of alpha(1,3)galactosyltransferase, showing that indeed the rat expresses two distinct alpha(1,3)galactosyltransferases, alpha(1,3)GT and iGb3 synthase. Rat alpha(1,3)galactosyltransferase shows a high amino acid sequence identity with the alpha(1,3)galactosyltransferase of mouse (90%), pig (76%), and ox (75%), in contrast to the low amino acid sequence identity (42%) with iGb3 synthase. The rat alpha(1,3)galactosyltransferase is expressed in heart, brain, spleen, kidney, and liver and has a similar intron/exon structure to the mouse alpha(1,3)galactosyltransferase. Transfection studies show that in contrast to the iGb3 synthase, rat alpha(1,3)galactosyltransferase can synthesize Galalpha(1,3)Gal on glycoproteins but cannot synthesize the glycolipid iGb3, defining two separate glycosylation pathways for the synthesis of Galalpha(1,3)Gal. Furthermore iGb3 synthase was found to be distinct from alpha(1,3)GT with its ability to synthesize poly-alpha-Gal glycolipid structures.


Assuntos
Galactosiltransferases/genética , Glicosiltransferases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Clonagem Molecular , Cricetinae , Dissacarídeos/análise , Dissacarídeos/biossíntese , Dissacarídeos/genética , Galactosiltransferases/biossíntese , Galactosiltransferases/metabolismo , Glicolipídeos/análise , Glicoproteínas/análise , Glicosilação , Glicosiltransferases/biossíntese , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
17.
Immunol Cell Biol ; 81(1): 79-85, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12534951

RESUMO

Cancer immunotherapy has traditionally undergone a 'revolution' every decade, from the use of Bacille Calmette-Guérin by scarification in the 1970s, to interleukin-2 therapies in the 1980s, and monoclonal antibody treatments in the early 1990s. Usually the early reports on the use of such agents were encouraging, but when more patients were studied in multiple centres, the initial promising results could not be confirmed. Now in a new century, we have more reagents and methods available than ever before - indeed, with such a plethora of reagents it is difficult to envisage them being fully and appropriately tested within the next decade, by which time there will be even more reagents to test. However, there have been three major advances which should lead to substantial progress in cancer immunotherapy: (1) the widespread use of genetic engineering, enabling identification of candidate vaccine proteins and manipulation of their sequences; (2) the production of antigens, antibodies and cytokines in large amounts by recombinant technologies, and (3) an understanding of the mode of presentation of peptides by major histocompatibility complex Class I and Class II molecules and their recognition by T cells. Despite these advances, there are major problems facing cancer immunotherapy, such as the ability of tumours to mutate and evade the immune system and the difficulty of precisely defining the interactions of effector cells in mediating 'rejection' or destruction of a tumour. There are clearly immunological similarities with diseases such as malaria and schistosomiasis, where the invading foreign organisms can use a variety of strategies to resist an elicited immune response. The failure to find a suitable vaccine for these diseases must lead to some pessimism for the development of immunotherapy for an autologous tumour. However, there are promising studies now in progress which should give an indication of the most important directions to follow. This review provides a commentary on aspects of cancer immunotherapy and in particular will deal with: (1) the selection of antigens as vaccine components; (2) the modes of presentation of antigens, particularly by major histocompatibility complex Class I molecules; and (3) new modes of delivery of vaccine immunogens.


Assuntos
Imunoterapia , Neoplasias/tratamento farmacológico , Apresentação de Antígeno/imunologia , Antígenos/imunologia , Citocinas/metabolismo , Bases de Dados Genéticas , Células Dendríticas , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imuno-Histoquímica , Linfócitos T Reguladores/metabolismo
18.
Xenotransplantation ; 10(6): 615-21, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14708531

RESUMO

We have previously reported that islets present in cultured fetal pig pancreas (FPP) are resistant to destruction by Galalpha(1,3)Gal antibodies and compliment, but are susceptible to the 'secondary' antibody response which occurs on transplanting pig islet tissue to Galo/o murine recipients. In a model of antibody-mediated graft rejection, we tested the resistance of porcine islets to antibody. Using FPP from human CD46 transgenic pigs, we now report that the complement regulator, CD46, affords protection from antibody-mediated rejection when mouse anti-pig serum (MAPS) was administered to scid mice bearing PFF grafts from either CD46 transgenic or normal pigs. Indeed, whereas normal pig islets were destroyed by an intraperitoneal (i.p.) injection of 0.1 to 0.2 ml of MAPS antibody, destruction of CD46-expressing transgenic islets required 0.5 ml, i.e. up to five times the amount. In contrast, there was no prolongation of the survival of CD46 transgenic mouse skin or heart major histocompatibility complex-compatible or -incompatible allografts--rejected by predominantly cellular immune mechanisms, as opposed to xenograft rejection. Although complement regulators have been examined for their protective role in hyperacute rejection of vascularized xenografts, it is clear that they also have protective effects in the later, antibody-mediated responses, but are unlikely to effect the inflammatory response in cell-mediated rejection.


Assuntos
Antígenos CD/imunologia , Transplante de Tecido Fetal , Rejeição de Enxerto , Ilhotas Pancreáticas/imunologia , Glicoproteínas de Membrana/imunologia , Transplante de Pâncreas , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Anticorpos Heterófilos/imunologia , Humanos , Insulina/metabolismo , Proteína Cofatora de Membrana , Camundongos , Camundongos SCID , Pâncreas/embriologia , Suínos
19.
Glycobiology ; 12(11): 729-39, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12460941

RESUMO

Historically, the most effective means of modifying cell surface carbohydrates has required the intracellular overexpression of glycosyltransferases or glycosidases and is dependent on the enzymes occupying a cellular localization close to the carbohydrate structures they modify. We report on relocalizing the lysosomal resident glycosidase human alpha-galactosidase to other regions of the cell, Golgi and cell surface, where it is in closer proximity for cleaving the carbohydrate structure Galalpha(1,3)Gal. Relocalization of alpha-galactosidase was achieved by using the transmembrane and cytoplasmic domains from the human protein furin, which is known to localize in the trans-Golgi network (TGN) and cell surface. Two chimeric forms of alpha-galactosidase were generated, one directing it to the TGN of the cell and the other to the cell surface, as shown by confocal microscopy. The relocalized enzymes have the ability to cleave terminal alpha-galactose as detected by expression on the cell surface. Furthermore, when expressed as a transgene in mice, the TGN form of alpha-galactosidase was more effective at decreasing cell surface terminal alpha-galactose than was the native lysosomal form. When expressed in conjunction with the alpha1,2fucosyltransferase that also decreases Galalpha(1,3)Gal, the reduction was additive. The ability to relocalize enzymes that modify cell surface carbohydrate structures has far-reaching implications in biology and may be useful in such fields as xenotransplantation and treatment of glycosidase disorders.


Assuntos
Membrana Celular/enzimologia , Galactose/metabolismo , alfa-Galactosidase/metabolismo , Rede trans-Golgi/metabolismo , Animais , Células COS , Furina/genética , Furina/metabolismo , Camundongos , Camundongos Transgênicos , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , alfa-Galactosidase/biossíntese , alfa-Galactosidase/genética , Rede trans-Golgi/enzimologia
20.
Chromosoma ; 111(2): 80-95, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12111331

RESUMO

The proto-oncogene pim-1 is a serine/threonine kinase the over-expression of which promotes lymphoma formation. Neither the normal function of Pim-1 nor the biochemical mechanism for cancer development mediated by the gene has been delineated, although recent studies have provided compelling evidence that Pim-1 is involved in differentiation and cell survival. We now provide the first evidence that Pim-1 may be involved in the proliferative process. By confocal microscopy, we observed a dynamic redistribution of Pim-1 during the cell cycle, the protein moving from the nucleus and cytoplasm in interphase to the spindle poles during mitosis. From a computer search for putative substrates of Pim-1 that are located in the spindle poles, we discovered that the nuclear mitotic apparatus (NuMA) protein has two peptide sequences that contain preferred phosphorylation sites for Pim-1 kinase. Recombinant glutathione-S-transferase-Pim-1 also readily phosphorylates immunoprecipitated NuMA. By confocal microscopy and co-immunoprecipitation we showed the interaction of the Pim-1 and NuMA proteins in HeLa cells that had been arrested during mitosis with nocodazole. Pim-1 also appeared to interact with heterochromatin-associated protein 1beta (HP1beta) and the cytoplasmic proteins dynein and dynactin via complex formation with NuMA. In our studies, overexpressed wild-type-Pim-1-GFP (green fluorescent protein) fusion protein was found to co-localize in the spindle pole with NuMA during mitosis. In contrast, the 'kinase-dead' mut-Pim-1-GFP fusion protein did not co-localize with NuMA, and appeared to promote apoptosis. Further evidence for apoptotic cell death was the observed blebbing and fragmentation of the chromosomes and a decrease in the level of NuMA protein detected by confocal microscopy. These results strongly suggest that Pim-1 kinase plays a role, most likely by phosphorylation, in promoting complex formation between NuMA, HP1beta, dynein and dynactin, a complex that is necessary for mitosis.


Assuntos
Mitose/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células 3T3 , Animais , Antígenos Nucleares , Sequência de Bases , Proteínas de Ciclo Celular , Homólogo 5 da Proteína Cromobox , Primers do DNA , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Camundongos , Microscopia Confocal , Mitose/efeitos dos fármacos , Nocodazol/farmacologia , Proteínas Associadas à Matriz Nuclear , Proteínas Nucleares/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/fisiologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-pim-1 , Proteínas Recombinantes de Fusão/metabolismo , Fuso Acromático , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...