Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Front Immunol ; 13: 1034774, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36405748

RESUMO

Tumor-targeting antibody (Ab)-fused cytokines, referred to as immunocytokines, are designed to increase antitumor efficacy and reduce toxicity through the tumor-directed delivery of cytokines. However, the poor localization and intratumoral penetration of immunocytokines, especially in solid tumors, pose a challenge to effectively stimulate antitumor immune cells to kill tumor cells within the tumor microenvironment. Here, we investigated the influence of the tumor antigen-binding kinetics of a murine interleukin 12 (mIL12)-based immunocytokine on tumor localization and diffusive intratumoral penetration, and hence the consequent antitumor activity, by activating effector T cells in immunocompetent mice bearing syngeneic colon tumors. Based on tumor-associated antigen HER2-specific Ab Herceptin (HCT)-fused mIL12 carrying one molecule of mIL12 (HCT-mono-mIL12 immunocytokine), we generated a panel of HCT-mono-mIL12 variants with different affinities (K D) mainly varying in their dissociation rates (k off) for HER2. Systemic administration of HCT-mono-mIL12 required an anti-HER2 affinity above a threshold (K D = 130 nM) for selective localization and antitumor activity to HER2-expressing tumors versus HER2-negative tumors. However, the high affinity (K D = 0.54 or 46 nM) due to the slow k off from HER2 antigen limited the depth of intratumoral penetration of HCT-mono-mIL12 and the consequent tumor infiltration of T cells, resulting in inferior antitumor activity compared with that of HCT-mono-mIL12 with moderate affinity of (K D = 130 nM) and a faster k off. The extent of intratumoral penetration of HCT-mono-mIL12 variants was strongly correlated with their tumor infiltration and intratumoral activation of CD4+ and CD8+ T cells to kill tumor cells. Collectively, our results demonstrate that when developing antitumor immunocytokines, tumor antigen-binding kinetics and affinity of the Ab moiety should be optimized to achieve maximal antitumor efficacy.


Assuntos
Neoplasias do Colo , Interleucina-12 , Camundongos , Animais , Linfócitos T CD8-Positivos , Trastuzumab , Receptor ErbB-2 , Citocinas , Antígenos de Neoplasias , Anticorpos , Microambiente Tumoral
3.
Biochem Biophys Res Commun ; 628: 18-24, 2022 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-36063598

RESUMO

VSIG4, a newly identified co-inhibitory molecule belonging to the B7-related family, is exclusively expressed on tissue-resident macrophages and is involved in the suppression of T cell proliferation and cytokine production. We sought to characterize the role of VSIG4 in anti-tumor immunity in the tumor microenvironment, focusing on VSIG4-expressing tumor-associated macrophages (TAMs). We found that VSIG4-expressing TAMs negatively regulated antigen-specific T cell proliferation and cytokine production through direct inhibition via cell cycle arrest, but not apoptosis, as well as through their arginase 1 activity. Furthermore, VSIG4-expressing TAMs suppress tumor-specific CD8+ T cell cytotoxicity. Therefore, our results suggest that VSIG4-expressing TAMs could be a negative cellular regulator of anti-tumor immunity in the tumor microenvironment.


Assuntos
Receptores de Complemento , Microambiente Tumoral , Macrófagos Associados a Tumor , Animais , Arginase/genética , Arginase/metabolismo , Citocinas/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Complemento/metabolismo , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo
4.
Mol Cancer ; 21(1): 102, 2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35459256

RESUMO

BACKGROUND: Redirecting pre-existing virus-specific cytotoxic CD8+ T lymphocytes (CTLs) to tumors by simulating a viral infection of the tumor cells has great potential for cancer immunotherapy. However, this strategy is limited by lack of amenable method for viral antigen delivery into the cytosol of target tumors. Here, we addressed the limit by developing a CD8+ T cell epitope-delivering antibody, termed a TEDbody, which was engineered to deliver a viral MHC-I epitope peptide into the cytosol of target tumor cells by fusion with a tumor-specific cytosol-penetrating antibody. METHODS: To direct human cytomegalovirus (CMV)-specific CTLs against tumors, we designed a series of TEDbodies carrying various CMV pp65 antigen-derived peptides. CMV-specific CTLs from blood of CMV-seropositive healthy donors were expanded for use in in vitro and in vivo experiments. Comprehensive cellular assays were performed to determine the presentation mechanism of TEDbody-mediated CMV peptide-MHC-I complex (CMV-pMHCI) on the surface of target tumor cells and the recognition and lysis by CMV-specific CTLs. In vivo CMV-pMHCI presentation and antitumor efficacy of TEDbody were evaluated in immunodeficient mice bearing human tumors. RESULTS: TEDbody delivered the fused epitope peptides into target tumor cells to be intracellularly processed and surface displayed in the form of CMV-pMHCI, leading to disguise target tumor cells as virally infected cells for recognition and lysis by CMV-specific CTLs. When systemically injected into tumor-bearing immunodeficient mice, TEDbody efficiently marked tumor cells with CMV-pMHCI to augment the proliferation and cytotoxic property of tumor-infiltrated CMV-specific CTLs, resulting in significant inhibition of the in vivo tumor growth by redirecting adoptively transferred CMV-specific CTLs. Further, combination of TEDbody with anti-OX40 agonistic antibody substantially enhanced the in vivo antitumor activity. CONCLUSION: Our study offers an effective technology for MHC-I antigen cytosolic delivery. TEDbody may thus have utility as a therapeutic cancer vaccine to redirect pre-existing anti-viral CTLs arising from previously exposed viral infections to attack tumors.


Assuntos
Infecções por Citomegalovirus , Neoplasias , Animais , Linfócitos T CD8-Positivos , Infecções por Citomegalovirus/terapia , Citosol , Epitopos , Humanos , Imunoterapia/métodos , Camundongos , Peptídeos , Linfócitos T Citotóxicos
5.
Int J Mol Sci ; 22(5)2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33652936

RESUMO

Human cytomegalovirus (CMV) infection is widespread among adults (60-90%) and is usually undetected in healthy individuals without symptoms but can cause severe diseases in immunocompromised hosts. T-cell receptor (TCR)-like antibodies (Abs), which recognize complex antigens (peptide-MHC complex, pMHC) composed of MHC molecules with embedded short peptides derived from intracellular proteins, including pathogenic viral proteins, can serve as diagnostic and/or therapeutic agents. In this study, we aimed to engineer a TCR-like Ab specific for pMHC comprising a CMV pp65 protein-derived peptide (495NLVPMVATV503; hereafter, CMVpp65495-503) in complex with MHC-I molecule human leukocyte antigen (HLA)-A*02:01 (CMVpp65495-503/HLA-A*02:01) to increase affinity by sequential mutagenesis of complementarity-determining regions using yeast surface display technology. Compared with the parental Ab, the final generated Ab (C1-17) showed ~67-fold enhanced binding affinity (KD ≈ 5.2 nM) for the soluble pMHC, thereby detecting the cell surface-displayed CMVpp65495-503/HLA-A*02:01 complex with high sensitivity and exquisite specificity. Thus, the new high-affinity TCR-like Ab may be used for the detection and treatment of CMV infection.


Assuntos
Anticorpos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Antígenos HLA-A/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas da Matriz Viral/imunologia , Afinidade de Anticorpos , Linhagem Celular , Humanos , Peptídeos/imunologia
6.
Front Immunol ; 11: 593748, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33488590

RESUMO

Patients with severe eosinophilic asthma (SEA; characterized by persistent eosinophilia in blood and airway tissues) experience frequent asthma exacerbations with poor clinical outcomes. Interleukin 5 (IL-5) and IL-5 receptor alpha subunit (IL-5α) play key roles in eosinophilia maintenance, and relevant therapeutic strategies include the development of antibodies (Abs) against IL-5 or IL-5α to control eosinophilia. Benralizumab, an anti-IL-5α Ab that depletes eosinophils mainly via Ab-dependent cell-mediated cytotoxicity and through blockage of IL-5 function on eosinophils, has been clinically approved for patients with SEA. Here, we report engineering of a new humanized anti-IL-5Rα Ab with potent biological activity. We first raised murine Abs against human IL-5Rα, humanized a leading murine Ab, and then further engineered the humanized Abs to enhance their affinity for IL-5Rα using the yeast surface display technology. The finally engineered version of the Ab, 5R65.7, with affinity (KD ≈ 4.64 nM) stronger than that of a clinically relevant benralizumab analogue (KD ≈ 26.8 nM) showed improved neutralizing activity toward IL-5-dependent cell proliferation in a reporter cell system. Domain level Ab epitope mapping revealed that 5R65.7 recognizes membrane-proximal domain 3 of IL-5Rα, distinct from domain I epitope of the benralizumab analogue. In ex vivo assays with peripheral eosinophils from patients with SEA and healthy donors, 5R65.7 manifested more potent biological activities than the benralizumab analogue did, including inhibition of IL-5-dependent proliferation of eosinophils and induction of eosinophil apoptosis through autologous natural-killer-cell-mediated Ab-dependent cell-mediated cytotoxicity. Our study provides a potent anti-IL-5Rα Ab, 5R65.7, which is worthy of further testing in preclinical and clinical trials against SEA as a potential alternative to the current therapeutic arsenal.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Subunidade alfa de Receptor de Interleucina-5/antagonistas & inibidores , Animais , Antiasmáticos/efeitos adversos , Antiasmáticos/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Asma/complicações , Asma/tratamento farmacológico , Asma/imunologia , Asma/metabolismo , Estudos de Casos e Controles , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Eosinófilos/metabolismo , Mapeamento de Epitopos/métodos , Epitopos/imunologia , Humanos , Imunização , Subunidade alfa de Receptor de Interleucina-5/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Camundongos , Proteínas Recombinantes
7.
Cancer Immunol Res ; 8(1): 46-56, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31554638

RESUMO

Regulatory T cells (Treg) are targeted for cancer immunotherapy because they suppress antitumor immunity. Although the importance of neuropilin-1 (NRP1) in the stability and function of intratumoral Tregs is well-documented, targeting of NRP1+ Tregs for anticancer immunotherapy has not been well explored. Here, we found that an NRP1 antagonist [Fc(AAG)-TPP11], generated by fusion of the NRP1-specific binding peptide TPP11 with the C-terminus of an effector function-deficient immunoglobulin Fc(AAG) variant, inhibits intratumoral NRP1+ Treg function and stability. Fc(AAG)-TPP11 triggered the internalization of NRP1, reducing its surface expression on Tregs and thereby inhibiting the suppressive function of Tregs. In two murine syngeneic tumor models, Fc(AAG)-TPP11 retarded tumor growth, comparable with a Treg-depleting anti-CTLA-4 antibody, without noticeable toxicity. Fc(AAG)-TPP11 inhibited NRP1-dependent Treg function, inducing unstable intratumoral Tregs, with reduced expression of Foxp3 and enhanced production of IFNγ, which subsequently increased the functionality and frequency of intratumoral CD8+ T cells. We also observed selective expression of NRP1 on Tregs isolated from human tumors, but not from the blood of healthy donors and patients with cancer, as well as ex vivo inhibition of intratumoral NRP1+ Treg function by Fc(AAG)-TPP11. Our results suggest that the NRP1 antagonist Fc(AAG)-TPP11 has therapeutic potential for the inhibition of intratumoral NRP1+ Tregs with limited unfavorable effects on peripheral Tregs.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Fragmentos Fc das Imunoglobulinas/química , Melanoma/imunologia , Neuropilina-1/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Feminino , Terapia de Imunossupressão , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Neuropilina-1/imunologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo , Linfócitos T Reguladores/metabolismo
8.
Sci Rep ; 9(1): 7772, 2019 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-31123339

RESUMO

Development of antagonistic antibody (Ab) against interleukin-4 receptor alpha (IL-4Rα) subunit of IL-4/IL-13 receptors is a promising therapeutic strategy for T helper 2 (TH2)-mediated allergic diseases such as asthma and atopic dermatitis. Here we isolated anti-human IL-4Rα antagonistic Abs from a large yeast surface-displayed human Ab library and further engineered their complementarity-determining regions to improve the affinity using yeast display technology, finally generating a candidate Ab, 4R34.1.19. When reformatted as human IgG1 form, 4R34.1.19 specifically bound to IL-4Rα with a high affinity (KD ≈ 178 pM) and effectively blocked IL-4- and IL-13-dependent signaling in a reporter cell system at a comparable level to that of the clinically approved anti-IL-4Rα dupilumab Ab analogue. Epitope mapping by alanine scanning mutagenesis revealed that 4R34.1.19 mainly bound to IL-4 binding sites on IL-4Rα with different epitopes from those of dupilumab analogue. Further, 4R34.1.19 efficiently inhibited IL-4-dependent proliferation of T cells among human peripheral blood mononuclear cells and suppressed the differentiation of naïve CD4+ T cells from healthy donors and asthmatic patients into TH2 cells, the activities of which were comparable to those of dupilumab analogue. Our work demonstrates that both affinity and epitope are critical factors for the efficacy of anti-IL-4Rα antagonistic Abs.


Assuntos
Anticorpos Monoclonais Humanizados , Asma/imunologia , Subunidade alfa de Receptor de Interleucina-4/imunologia , Leucócitos Mononucleares/imunologia , Adulto , Humanos
9.
Oncoimmunology ; 7(7): e1438800, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29900039

RESUMO

Interleukin-12 (IL12) (p35/p40 complex) is a heterodimeric cytokine with potent anti-tumor activity. However, its short serum half-life and high dose-related toxicities limit its clinical efficacy. Here, we constructed heterodimeric immunoglobulin Fc-fused mouse IL12 (mIL12) in a monovalent binding format (mono-mIL12-Fc) to generate long-acting mIL12 in the naturally occurring heterodimeric form. Mono-mIL12-Fc exhibited a much longer plasma half-life than recombinant mIL12, enabling twice-weekly systemic injections to remove established tumors in syngeneic mouse models. Mono-mIL12-Fc was more potent than wild-type Fc-based bivalent-binding IL12-Fc (bi-mIL12-Fc) for eradicating large established immunogenic tumors without noticeable toxicities by enhancing interferon-γ production and the proliferation of immune effector cells in tumors. More importantly, mono-mIL12-Fc triggered weaker IL12 signaling than bi-mIL12-Fc, favoring the generation of functional and protective memory CD8+ T cells. Our results demonstrate that heterodimeric-Fc-fused IL12 is a suitable format for augmenting adaptive CD8+ T cell immune responses, providing a practical alternative to the systemic administration of IL12 for antitumor therapy.

10.
Nat Commun ; 8: 15090, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28489072

RESUMO

Oncogenic Ras mutants, frequently detected in human cancers, are high-priority anticancer drug targets. However, direct inhibition of oncogenic Ras mutants with small molecules has been extremely challenging. Here we report the development of a human IgG1 format antibody, RT11, which internalizes into the cytosol of living cells and selectively binds to the activated GTP-bound form of various oncogenic Ras mutants to block the interactions with effector proteins, thereby suppressing downstream signalling and exerting anti-proliferative effects in a variety of tumour cells harbouring oncogenic Ras mutants. When systemically administered, an RT11 variant with an additional tumour-associated integrin binding moiety for tumour tissue targeting significantly inhibits the in vivo growth of oncogenic Ras-mutated tumour xenografts in mice, but not wild-type Ras-harbouring tumours. Our results demonstrate the feasibility of developing therapeutic antibodies for direct targeting of cytosolic proteins that are inaccessible using current antibody technology.


Assuntos
Anticorpos Monoclonais/farmacologia , Proliferação de Células/efeitos dos fármacos , Citosol/metabolismo , Imunoglobulina G/farmacologia , Neoplasias/genética , Proteínas ras/genética , Animais , Linhagem Celular Tumoral , GTP Fosfo-Hidrolases/antagonistas & inibidores , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Células HL-60 , Células HT29 , Células HeLa , Humanos , Células K562 , Células MCF-7 , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Células NIH 3T3 , Transplante de Neoplasias , Neoplasias/tratamento farmacológico , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/antagonistas & inibidores , Proteínas ras/metabolismo
11.
Immunol Lett ; 165(2): 78-83, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25929804

RESUMO

VSIG4 acts as a co-inhibitory ligand to negatively regulate T cell proliferation and cytokine production, and its expression is restricted to macrophages. We hypothesized that endogenous VSIG4 impairs helper T cell functions and then inhibits the subsequent antibody response. Isotype switching of ovalbumin (OVA)-specific antibody subclasses to IgG1, IgG2a, IgG2b, and IgG3 was enhanced in OVA-immunized VSIG4 knockout (KO) mice. 2,4,6-Trinitrophenyl hapten (TNP) - Keyhole Limpet Hemocyanin (KLH)-primed B cells cocultured with OVA-primed CD4(+) T cells from OVA-immunized VSIG4 KO mice in the presence of TNP-OVA showed enhanced isotype switching to IgG subclasses compared to those cocultured with cells isolated from OVA-immunized wild-type (WT) mice. Furthermore, the levels of CD40L expression, the frequency of memory CD4(+) T cells, and the production of isotype switching-inducing cytokines increased significantly in OVA-primed CD4(+) T cells from VSIG4 KO mice. T cells from OVA-specific T cell receptor (TCR) transgenic mice produced more IFN-γ when cocultured with macrophages from VSIG4 KO mice compared to WT mice. Thus, our results demonstrate that macrophage-associated VSIG4 plays a negative role in helper T cell-dependent isotype switching by inhibiting helper T cell activation and differentiation, and suppressing the isotype switching-inducing cytokine production in antigen-primed CD4(+) helper T cells.


Assuntos
Macrófagos/fisiologia , Receptores de Complemento/metabolismo , Linfócitos T Auxiliares-Indutores/fisiologia , Animais , Formação de Anticorpos/genética , Ligante de CD40/metabolismo , Diferenciação Celular/genética , Técnicas de Cocultura , Citocinas/metabolismo , Células HEK293 , Humanos , Switching de Imunoglobulina/genética , Memória Imunológica/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento/genética
12.
Immune Netw ; 13(5): 184-93, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24198743

RESUMO

Co-signaling molecules are surface glycoproteins that positively or negatively regulate the T cell response to antigen. Co-signaling ligands and receptors crosstalk between the surfaces of antigen-presenting cells (APCs) and T cells, and modulate the ultimate magnitude and quality of T cell receptor (TCR) signaling. In the past 10 years, the field of co-signaling research has been advanced by the understanding of underlying mechanisms of the immune modulation led by newly identified co-signaling molecules and the successful preclinical and clinical trials targeting co-inhibitory molecules called immune checkpoints in the treatment of autoimmune diseases and cancers. In this review, we briefly describe the characteristics of well-known B7 co-signaling family members regarding the expression, functions and therapeutic implications and to introduce newly identified B7 members such as B7-H5, B7-H6, and B7-H7.

13.
Immunol Lett ; 156(1-2): 132-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24148970

RESUMO

Tumor cells have been used as the tumor antigen sources for developing cancer vaccines. Due to their low immunogenicity, tumor antigens are combined with various adjuvants to enhance immunogenicity of cancer vaccines. Among them, a natural killer T cell (NKT)-ligand, α-galactosylceramide (αGC) has been reported as a powerful adjuvant showing therapeutic effects in solid tumors as well as hematological malignancies including lymphoma. In this study, we applied αGC-based tumor cell vaccine in mouse multiple myeloma model. The αGC-loaded MOPC315BM myeloma cell vaccine efficiently retarded tumor growth, induced regression of established tumors, and protected surviving mice from tumor rechallenge. Therapeutic responses were associated with induction of strong humoral immune responses, including myeloma-specific antibodies, and cellular immune responses, including myeloma-specific CD8(+) cytotoxic T lymphocytes and memory T cells. In addition, regulatory T cells were significantly decreased in mice that received the αGC-loaded myeloma cell vaccine. Thus, our results demonstrated that αGC-loaded myeloma vaccine efficiently promoted NKT-dependent anti-tumor immunity in a mouse model. These findings are informative for improving the efficacy of tumor-cell-based immunotherapy for patients with MM and other CD1d-expressing tumors.


Assuntos
Vacinas Anticâncer/imunologia , Galactosilceramidas/imunologia , Mieloma Múltiplo/imunologia , Células T Matadoras Naturais/imunologia , Animais , Anticorpos/sangue , Anticorpos/imunologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Testes Imunológicos de Citotoxicidade/métodos , Citotoxicidade Imunológica/imunologia , Citometria de Fluxo , Imunoterapia/métodos , Interferon gama/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Células T Matadoras Naturais/metabolismo , Baço/citologia , Baço/imunologia , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/metabolismo , Resultado do Tratamento , Vacinação/métodos
14.
Hepatology ; 56(5): 1838-48, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22711680

RESUMO

UNLABELLED: V-set and Ig domain-containing 4 (VSIG4, CRIg, or Z39Ig), a newly identified B7-related cosignaling molecule, is a complement receptor and a coinhibitory ligand that negatively regulates T-cell immunity. Despite its exclusive expression on liver Kupffer cells (KCs) that play key roles in liver tolerance, the physiological role of VSIG4 in liver tolerance remains undefined. Mice lacking VSIG4 had poor survival rates and severe liver pathology in a concanavalin A (ConA)-induced hepatitis (CIH) model, which could be prevented by adoptive transfer of VSIG4(+) KCs. The absence of VSIG4 rendered endogenous liver T- and natural killer T (NKT)-cells more responsive to antigen-specific stimulation and impaired tolerance induction in those cells against their cognate antigens. T-cell costimulation with VSIG4.Ig suppressed Th1-, Th2-, and Th17-type cytokine production and arrested the cell cycle at the G(0) /G(1) phase but did not induce apoptosis in vitro. VSIG4-mediated tolerance induction and cell-cycle arrest were further supported by down-regulation of G(1) phase-specific Cdk2, Cdk4, and Cdk6, and up-regulation of tolerance-inducing p27(KIP-1) in VSIG4.Ig-stimulated T-cells. Administration of soluble VSIG4.Ig to wildtype mice prevented CIH development and prolonged the survival of mice with established CIH. CONCLUSION: Collectively, our results suggest that VSIG4(+) KCs play a critical role in the induction and maintenance of liver T- and NKT-cell tolerance, and that modulation of the VSIG4 pathway using a VSIG4.Ig fusion protein may provide useful immunological therapies against immune-mediated liver injury including autoimmune hepatitis.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/imunologia , Tolerância Imunológica , Células de Kupffer/imunologia , Fígado/imunologia , Fígado/lesões , Receptores de Complemento/imunologia , Linfócitos T/imunologia , Animais , Proliferação de Células , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/genética , Técnicas de Cocultura , Concanavalina A/toxicidade , Citocinas/metabolismo , Galactosilceramidas/imunologia , Hepatite/imunologia , Imunoglobulina G/imunologia , Células de Kupffer/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Ovalbumina/imunologia , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/metabolismo
15.
J Virol ; 86(14): 7616-24, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22573867

RESUMO

Phylogenetic relatedness and cocirculation of several major human pathogen flaviviruses are recognized as a possible cause of deleterious immune responses to mixed infection or immunization and call for a greater understanding of the inter-Flavivirus protein homologies. This study focused on the identification of human leukocyte antigen (HLA)-restricted West Nile virus (WNV) T-cell ligands and characterization of their distribution in reported sequence data of WNV and other flaviviruses. H-2-deficient mice transgenic for either A2, A24, B7, DR2, DR3, or DR4 HLA alleles were immunized with overlapping peptides of the WNV proteome, and peptide-specific T-cell activation was measured by gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) assays. Approximately 30% (137) of the WNV proteome peptides were identified as HLA-restricted T-cell ligands. The majority of these ligands were conserved in ∼≥88% of analyzed WNV sequences. Notably, only 51 were WNV specific, and the remaining 86, chiefly of E, NS3, and NS5, shared an identity of nine or more consecutive amino acids with sequences of 64 other flaviviruses, including several major human pathogens. Many of the shared ligands had an incidence of >50% in the analyzed sequences of one or more of six major flaviviruses. The multitude of WNV sequences shared with other flaviviruses as interspecies variants highlights the possible hazard of defective T-cell activation by altered peptide ligands in the event of dual exposure to WNV and other flaviviruses, by either infection or immunization. The data suggest the possible preferred use of sequences that are pathogen specific with minimum interspecies sequence homology for the design of Flavivirus vaccines.


Assuntos
Antígenos Virais/imunologia , Flavivirus/imunologia , Antígenos de Histocompatibilidade/imunologia , Ativação Linfocitária , Linfócitos T/imunologia , Proteínas Virais/imunologia , Vírus do Nilo Ocidental/imunologia , Sequência de Aminoácidos , Animais , ELISPOT , Variação Genética , Antígenos de Histocompatibilidade/genética , Interferon gama , Ligantes , Camundongos , Camundongos Transgênicos , Proteoma , Linfócitos T/metabolismo , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/metabolismo
16.
Vaccine ; 29(1): 34-44, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20974308

RESUMO

Tumor microenvironment has emerged as one of the major obstacles against the clinical efficacy of dendritic cell (DC) vaccines. Tumor-derived IL-6 may inhibit the differentiation of hematopoietic progenitor cells into DCs and suppress DC maturation, rendering DCs tolerogenic. We hypothesized that silencing the IL-6 receptor alpha chain (IL-6Rα) would restore the functional competence of DC vaccines in mice with an IL-6-producing TC-1 tumor, and eventually give rise to protective immunity. We found that the IL-6Rα knockdown-DC vaccine significantly enhanced the frequency of tumor-specific CD8(+) CTLs-producing effector molecules such as IFN-γ, TNF-α, FasL, perforin, and granzyme B, and generated more CD8(+) memory T cells, leading to the substantially prolonged survival of TC-1 tumor-bearing mice.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Técnicas de Silenciamento de Genes , Subunidade alfa de Receptor de Interleucina-6/biossíntese , Interleucina-6/metabolismo , Neoplasias/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia
17.
PLoS One ; 5(1): e8574, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-20052293

RESUMO

This report describes the identification and bioinformatics analysis of HLA-DR4-restricted HIV-1 Gag epitope peptides, and the application of dendritic cell mediated immunization of DNA plasmid constructs. BALB/c (H-2d) and HLA-DR4 (DRA1*0101, DRB1*0401) transgenic mice were immunized with immature dendritic cells transfected by a recombinant DNA plasmid encoding the lysosome-associated membrane protein-1/HIV-1 Gag (pLAMP/gag) chimera antigen. Three immunization protocols were compared: 1) primary subcutaneous immunization with 1x10(5) immature dendritic cells transfected by electroporation with the pLAMP/gag DNA plasmid, and a second subcutaneous immunization with the naked pLAMP/gag DNA plasmid; 2) primary immunization as above, and a second subcutaneous immunization with a pool of overlapping peptides spanning the HIV-1 Gag sequence; and 3) immunization twice by subcutaneous injection of the pLAMP/gag DNA plasmid. Primary immunization with pLAMP/gag-transfected dendritic cells elicited the greatest number of peptide specific T-cell responses, as measured by ex vivo IFN-gamma ELISpot assay, both in BALB/c and HLA-DR4 transgenic mice. The pLAMP/gag-transfected dendritic cells prime and naked DNA boost immunization protocol also resulted in an increased apparent avidity of peptide in the ELISpot assay. Strikingly, 20 of 25 peptide-specific T-cell responses in the HLA-DR4 transgenic mice contained sequences that corresponded, entirely or partially to 18 of the 19 human HLA-DR4 epitopes listed in the HIV molecular immunology database. Selection of the most conserved epitope peptides as vaccine targets was facilitated by analysis of their representation and variability in all reported sequences. These data provide a model system that demonstrates a) the superiority of immunization with dendritic cells transfected with LAMP/gag plasmid DNA, as compared to naked DNA, b) the value of HLA transgenic mice as a model system for the identification and evaluation of epitope-based vaccine strategies, and c) the application of variability analysis across reported sequences in public databases for selection of historically conserved HIV epitopes as vaccine targets.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , DNA/administração & dosagem , Células Dendríticas/metabolismo , Epitopos/imunologia , Produtos do Gene gag/genética , Antígeno HLA-DR4/imunologia , Plasmídeos , Sequência de Aminoácidos , Animais , Células Cultivadas , Eletroporação , Ensaio de Imunoadsorção Enzimática , Epitopos/química , Proteínas Ligadas por GPI , HIV-1/imunologia , Antígeno HLA-DR4/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Dados de Sequência Molecular
18.
Immunol Lett ; 127(1): 39-47, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19723542

RESUMO

While programmed death-1 (PD-1), a co-inhibitory member of CD28 immunoglobulin superfamily plays negative roles in effector functions of T cells and B cells, little is known about the function of PD-1 expressed on innate immune cells. In this study, we demonstrate that IL-12 production was greatly suppressed in LPS-stimulated RAW264.7 cells upon PD-1 engagement with B7-H1.Fc fusion protein, and was restored in the presence of antagonistic anti-PD-1 mAb. PD-1-mediated suppression of IL-12 production in LPS-stimulated RAW264.7 cells was mediated by inhibition of Janus N-terminal-linked kinase (JNK) signaling pathway, and to a lesser extent, phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway through the recruitment of SHP-2 to PD-1 cytoplasmic tail. B7-H1.Fc-mediated PD-1 engagement also downregulates the expression of co-stimulatory molecules such as CD80, CD86, MHC class I and II proteins in LPS-stimulated RAW264.7 cells. Furthermore, the endocytic activity is enhanced but the allostimulatory capacity is suppressed in LPS-treated RAW264.7 cells upon PD-1 engagement. Taken together, our results reveal a novel function of macrophage PD-1 in the negative regulation of IL-12 synthesis and differentiation into dendritic cell-like cells.


Assuntos
Interleucina-12/biossíntese , Macrófagos/metabolismo , Receptores Imunológicos/metabolismo , Animais , Anticorpos Bloqueadores , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Antígeno B7-1/metabolismo , Antígeno B7-1/farmacologia , Antígeno B7-H1 , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Linhagem Celular , Regulação para Baixo , Endocitose/efeitos dos fármacos , Interleucina-12/genética , Lipopolissacarídeos/farmacologia , Teste de Cultura Mista de Linfócitos , MAP Quinase Quinase 4/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacologia , Camundongos , Peptídeos/metabolismo , Peptídeos/farmacologia , Receptor de Morte Celular Programada 1 , Receptores Imunológicos/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
19.
PLoS One ; 4(4): e5352, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19401763

RESUMO

West Nile virus (WNV) has emerged globally as an increasingly important pathogen for humans and domestic animals. Studies of the evolutionary diversity of the virus over its known history will help to elucidate conserved sites, and characterize their correspondence to other pathogens and their relevance to the immune system. We describe a large-scale analysis of the entire WNV proteome, aimed at identifying and characterizing evolutionarily conserved amino acid sequences. This study, which used 2,746 WNV protein sequences collected from the NCBI GenPept database, focused on analysis of peptides of length 9 amino acids or more, which are immunologically relevant as potential T-cell epitopes. Entropy-based analysis of the diversity of WNV sequences, revealed the presence of numerous evolutionarily stable nonamer positions across the proteome (entropy value of < or = 1). The representation (frequency) of nonamers variant to the predominant peptide at these stable positions was, generally, low (< or = 10% of the WNV sequences analyzed). Eighty-eight fragments of length 9-29 amino acids, representing approximately 34% of the WNV polyprotein length, were identified to be identical and evolutionarily stable in all analyzed WNV sequences. Of the 88 completely conserved sequences, 67 are also present in other flaviviruses, and several have been associated with the functional and structural properties of viral proteins. Immunoinformatic analysis revealed that the majority (78/88) of conserved sequences are potentially immunogenic, while 44 contained experimentally confirmed human T-cell epitopes. This study identified a comprehensive catalogue of completely conserved WNV sequences, many of which are shared by other flaviviruses, and majority are potential epitopes. The complete conservation of these immunologically relevant sequences through the entire recorded WNV history suggests they will be valuable as components of peptide-specific vaccines or other therapeutic applications, for sequence-specific diagnosis of a wide-range of Flavivirus infections, and for studies of homologous sequences among other flaviviruses.


Assuntos
Proteínas Virais/genética , Vírus do Nilo Ocidental/genética , Sequência de Aminoácidos , Animais , Antígenos Virais/genética , Sequência Conservada , Bases de Dados de Proteínas , Epitopos de Linfócito T/genética , Evolução Molecular , Variação Genética , Antígenos HLA , Humanos , Dados de Sequência Molecular , Proteoma , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade
20.
J Med Food ; 11(4): 717-22, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19053865

RESUMO

The kind of salt used in doenjang preparation is one of the most important factors that affect the chemopreventive activities of doenjang. Bamboo salt (BS) is known to exert various therapeutic effects on several diseases. In this study, antimutagenic and anticlastogenic effects of methanol extracts of doenjang made with either one-time heat-treated BS (BS1-D), nine-times heat-treated BS (BS9-D), sun-dried salt (SDS-D), or purified salt (PS-D) using the Ames test and micronucleus test. BS1-D and BS9-D showed higher inhibitory effects than PS-D and SDS-D on mutagenicity induced by aflatoxin B(1), N-methyl-N'-nitro-N-nitrosoguanidine, and 4-nitroquinoline-1-oxide. BS1-D exerted more suppressive effects on chromosome aberration than SDS-D on mitomycin C-induced micronucleus in mice. It can be concluded that the chemopreventive effect of doenjang was significantly increased by using BS instead of the more typical commercial salts, SDS and PS (P < .05). This is probably due to the changes in the chemical and physical properties of the salt, especially increased content of K and P, during the processing of BS and to differences in fermented products formed during the doenjang fermentation in the presence of BS.


Assuntos
Antimutagênicos/farmacologia , Mutagênese/efeitos dos fármacos , Cloreto de Sódio na Dieta/farmacologia , Alimentos de Soja , Animais , Aberrações Cromossômicas/efeitos dos fármacos , Fermentação , Coreia (Geográfico) , Masculino , Camundongos , Testes para Micronúcleos , Minerais/análise , Mutagênicos/farmacologia , Poaceae , Cloreto de Sódio na Dieta/administração & dosagem , Glycine max/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...