Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
1.
Neurotherapeutics ; 18(3): 1980-1994, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33829411

RESUMO

Numerous therapies aimed at driving an effective anti-glioma response have been employed over the last decade; nevertheless, survival outcomes for patients remain dismal. This may be due to the expression of immune-checkpoint ligands such as PD-L1 by glioblastoma (GBM) cells which interact with their respective receptors on tumor-infiltrating effector T cells curtailing the activation of anti-GBM CD8+ T cell-mediated responses. Therefore, a combinatorial regimen to abolish immunosuppression would provide a powerful therapeutic approach against GBM. We developed a peptide ligand (CD200AR-L) that binds an uncharacterized CD200 immune-checkpoint activation receptor (CD200AR). We sought to test the hypothesis that CD200AR-L/CD200AR binding signals via he DAP10&12 pathways through in vitro studies by analyzing transcription, protein, and phosphorylation, and in vivo loss of function studies using inhibitors to select signaling molecules. We report that CD200AR-L/CD200AR binding induces an initial activation of the DAP10&12 pathways followed by a decrease in activity within 30 min, followed by reactivation via a positive feedback loop. Further in vivo studies using DAP10&12KO mice revealed that DAP10, but not DAP12, is required for tumor control. When we combined CD200AR-L with an immune-stimulatory gene therapy, in an intracranial GBM model in vivo, we observed increased median survival, and long-term survivors. These studies are the first to characterize the signaling pathway used by the CD200AR, demonstrating a novel strategy for modulating immune checkpoints for immunotherapy currently being analyzed in a phase I adult trial.


Assuntos
Antígenos CD/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Inibidores de Checkpoint Imunológico/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/deficiência , Sequência de Aminoácidos , Animais , Antígenos CD/administração & dosagem , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Feminino , Terapia Genética/métodos , Glioma/tratamento farmacológico , Glioma/genética , Inibidores de Checkpoint Imunológico/administração & dosagem , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Terciária de Proteína , Receptores Imunológicos/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/fisiologia
2.
Immunol Res ; 68(3): 141-151, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32529460

RESUMO

Aberrant T helper-2 (Th2) responses play a critical role in the pathogenesis of allergic diseases. The underlying mechanism is to be further investigated. It is reported that soluble CD83 (sCD83) has immune-regulatory effects. This study aims to investigate the role of sCD83 in the regulation of Th2 polarization. Blood samples were collected from pediatric patients with food allergy (FA). The Th2 response was analyzed by pertinent immunological approaches. An FA murine model was developed to test the role of sCD83 in the regulation of FA response. We found that the serum sCD83 levels were lower in FA patients. A negative correlation was detected between serum sCD83 levels and serum Th2 cytokine levels. The presence of sCD83 suppressed Th2 cell differentiation and antigen-specific Th2 cell activation. sCD83 upregulated the T-bet expression and suppressed the GATA3 expression in CD4+ T cells. Administration of sCD83 suppressed experimental FA. Pediatric FA patients have low serum sCD83 levels. Administration of sCD83 can alleviate experimental FA via suppression of aberrant Th2 polarization.


Assuntos
Antígenos CD/metabolismo , Hipersensibilidade a Ovo/imunologia , Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Células Th2/imunologia , Adolescente , Animais , Antígenos CD/administração & dosagem , Antígenos CD/sangue , Células Cultivadas , Criança , Modelos Animais de Doenças , Hipersensibilidade a Ovo/sangue , Hipersensibilidade a Ovo/tratamento farmacológico , Feminino , Fator de Transcrição GATA3/metabolismo , Regulação da Expressão Gênica/imunologia , Humanos , Imunoglobulinas/administração & dosagem , Imunoglobulinas/sangue , Ativação Linfocitária , Masculino , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/sangue , Ovalbumina/efeitos adversos , Cultura Primária de Células , Proteínas com Domínio T/metabolismo , Antígeno CD83
3.
J Immunother Cancer ; 8(1)2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32217757

RESUMO

BACKGROUND: CD6 is a lymphocyte surface co-receptor physically associated with the T-cell receptor (TCR)/CD3 complex at the center of the immunological synapse. There, CD6 assists in cell-to-cell contact stabilization and modulation of activation/differentiation events through interaction with CD166/ALCAM (activated leukocyte cell adhesion molecule), its main reported ligand. While accumulating evidence is attracting new interest on targeting CD6 for therapeutic purposes in autoimmune disorders, little is known on its potential in cancer. In an attempt to elucidate the in vivo relevance of blocking CD6-mediated interactions in health and disease, we explored the consequences of expressing high circulating levels of a soluble form CD6 (sCD6) as a decoy receptor. METHODS: High sCD6 serum levels were achieved by using transgenic C57BL/6 mice expressing human sCD6 under the control of lymphoid-specific transcriptional elements (shCD6LckEµTg) or wild type either transduced with hepatotropic adeno-associated virus coding for mouse sCD6 or undergoing repeated infusions of recombinant human sCD6 protein. Characterization of sCD6-induced changes was performed by ex vivo flow cytometry and functional analyses of mouse lymphoid organ cells. The in vivo relevance of those changes was explored by challenging mice with subcutaneous or metastatic tumors induced by syngeneic cancer cells of different lineage origins. RESULTS: Through a combination of in vitro and in vivo studies, we show that circulating sCD6 expression induces defective regulatory T cell (Treg) generation and function, decreased CD166/ALCAM-mediated tumor cell proliferation/migration and impaired galectin-induced T-cell apoptosis, supporting the fact that sCD6 modulates antitumor lymphocyte effector function and tumorigenesis. Accordingly, sCD6 expression in vivo resulted in delayed subcutaneous tumor growth and/or reduced metastasis on challenge of mice with syngeneic cancer cells. CONCLUSIONS: Evidence is provided for the disruption of CD6 receptor-ligand interactions as a feasible immunomodulatory approach in cancer.


Assuntos
Antígenos CD/sangue , Antígenos de Diferenciação de Linfócitos T/sangue , Neoplasias Pulmonares/imunologia , Linfoma de Células T/imunologia , Melanoma Experimental/imunologia , Sarcoma Experimental/imunologia , Linfócitos T Reguladores/imunologia , Molécula de Adesão de Leucócito Ativado/imunologia , Molécula de Adesão de Leucócito Ativado/metabolismo , Animais , Antígenos CD/administração & dosagem , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/administração & dosagem , Antígenos de Diferenciação de Linfócitos T/biossíntese , Antígenos de Diferenciação de Linfócitos T/genética , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Linfoma de Células T/metabolismo , Masculino , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Proteínas Recombinantes/genética , Sarcoma Experimental/sangue , Sarcoma Experimental/patologia , Linfócitos T Reguladores/metabolismo
4.
J Virol ; 93(19)2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31315995

RESUMO

CD69 is highly expressed on the leukocyte surface upon viral infection, and its regulatory role in the vaccinia virus (VACV) immune response has been recently demonstrated using CD69-/- mice. Here, we show augmented control of VACV infection using the anti-human CD69 monoclonal antibody (MAb) 2.8 as both preventive and therapeutic treatment for mice expressing human CD69. This control was related to increased natural killer (NK) cell reactivity and increased numbers of cytokine-producing T and NK cells in the periphery. Moreover, similarly increased immunity and protection against VACV were reproduced over both long and short periods in anti-mouse CD69 MAb 2.2-treated immunocompetent wild-type (WT) mice and immunodeficient Rag2-/- CD69+/+ mice. This result was not due to synergy between infection and anti-CD69 treatment since, in the absence of infection, anti-human CD69 targeting induced immune activation, which was characterized by mobilization, proliferation, and enhanced survival of immune cells as well as marked production of several innate proinflammatory cytokines by immune cells. Additionally, we showed that the rapid leukocyte effect induced by anti-CD69 MAb treatment was dependent on mTOR signaling. These properties suggest the potential of CD69-targeted therapy as an antiviral adjuvant to prevent derived infections.IMPORTANCE In this study, we demonstrate the influence of human and mouse anti-CD69 therapies on the immune response to VACV infection. We report that targeting CD69 increases the leukocyte numbers in the secondary lymphoid organs during infection and improves the capacity to clear the viral infection. Targeting CD69 increases the numbers of gamma interferon (IFN-γ)- and tumor necrosis factor alpha (TNF-α)-producing NK and T cells. In mice expressing human CD69, treatment with an anti-CD69 MAb produces increases in cytokine production, survival, and proliferation mediated in part by mTOR signaling. These results, together with the fact that we have mainly worked with a human-CD69 transgenic model, reveal CD69 as a treatment target to enhance vaccine protectiveness.


Assuntos
Fatores Imunológicos/antagonistas & inibidores , Células Matadoras Naturais/imunologia , Lectinas Tipo C/antagonistas & inibidores , Linfócitos T/imunologia , Vaccinia virus/imunologia , Vacínia/prevenção & controle , Animais , Anticorpos Monoclonais/administração & dosagem , Antígenos CD/administração & dosagem , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos T/administração & dosagem , Antígenos de Diferenciação de Linfócitos T/genética , Modelos Animais de Doenças , Humanos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/genética , Lectinas Tipo C/administração & dosagem , Lectinas Tipo C/genética , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Vacínia/imunologia , Vacínia/terapia
5.
J Exp Clin Cancer Res ; 38(1): 168, 2019 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-30995926

RESUMO

BACKGROUND: Chimeric antigen receptor (CAR)-engineered T cells have displayed outstanding performance in the treatment of patients with hematological malignancies. However, their efficacy against solid tumors has been largely limited. METHODS: In this study, human osteosarcoma cell lines were prepared, flow cytometry using antibodies against CD166 was performed on different cell samples. CD166-specific T cells were obtained by viral gene transfer of corresponding DNA plasmids and selectively expanded using IL-2 and IL-15. The ability of CD166.BBζ CAR-T cells to kill CD166+ osteosarcoma cells was evaluated in vitro and in vivo. RESULTS: CD166 was selectively expressed on four different human osteosarcoma cell lines, indicating its role as the novel target for CAR-T cell therapy. CD166.BBζ CAR-T cells killed osteosarcoma cell lines in vitro; the cytotoxicity correlated with the level of CD166 expression on the tumor cells. Intravenous injection of CD166.BBζ CAR-T cells into mice resulted in the regression of the tumor with no obvious toxicity. CONCLUSIONS: Together, the data suggest that CD166.BBζ CAR-T cells may serve as a new therapeutic strategy in the future clinical practice for the treatment of osteosarcoma.


Assuntos
Antígenos CD/administração & dosagem , Moléculas de Adesão Celular Neuronais/administração & dosagem , Proteínas Fetais/administração & dosagem , Imunoterapia Adotiva/métodos , Osteossarcoma/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Ligante 4-1BB/administração & dosagem , Ligante 4-1BB/genética , Ligante 4-1BB/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas Fetais/genética , Proteínas Fetais/imunologia , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-15/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Camundongos , Osteossarcoma/genética , Osteossarcoma/imunologia , Osteossarcoma/patologia , Receptores de Antígenos de Linfócitos T/administração & dosagem , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/administração & dosagem , Receptores de Antígenos Quiméricos/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Future Oncol ; 15(17): 1963-1973, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30977393

RESUMO

Eftilagimod alpha (IMP321), a soluble dimeric recombinant form of LAG-3, is a first-in-class antigen presenting cell activator under clinical development. By stimulating dendritic cells through MHC class II molecules, IMP321 was proven to induce sustained immune responses. Combining active immunotherapy with a standard cytotoxic chemotherapy regimen represents a promising novel strategy that might lead to therapeutic improvements in metastatic breast cancer. Here, we describe the rationale and design of AIPAC (NCT02614833), a double-blind, randomized, multicenter Phase IIb study evaluating IMP321 plus paclitaxel as a first-line chemotherapy compared with paclitaxel plus placebo in hormone receptor-positive metastatic breast cancer patients. The primary end point is progression-free survival and key secondary objectives include overall survival, safety, quality of life and objective response rate.


Assuntos
Antígenos CD/administração & dosagem , Antineoplásicos Imunológicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Paclitaxel/administração & dosagem , Adulto , Antígenos CD/efeitos adversos , Antineoplásicos Imunológicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Ensaios Clínicos Fase II como Assunto , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Feminino , Humanos , Estudos Multicêntricos como Assunto , Paclitaxel/efeitos adversos , Placebos/administração & dosagem , Placebos/efeitos adversos , Intervalo Livre de Progressão , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteína do Gene 3 de Ativação de Linfócitos
7.
Am J Hum Genet ; 104(2): 341-347, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30712775

RESUMO

Erythropoietic protoporphyria (EPP) is a hereditary disease characterized by a deficiency in ferrochelatase (FECH) activity. FECH activity is responsible for the accumulation of protoporphyrin IX (PPIX). Without etiopathogenic treatment, EPP manifests as severe photosensitivity. 95% of affected individuals present a hypomorphic FECH allele trans to a loss-of-function (LOF) FECH mutation, resulting in a reduction in FECH activity in erythroblasts below a critical threshold. The hypomorphic allele promotes the use of a cryptic acceptor splice site, generating an aberrant FECH mRNA, which is responsible for the reduced level of wild-type FECH mRNA and, ultimately, FECH activity. We have previously identified an antisense oligonucleotide (AON), AON-V1 (V1), that redirects splicing to the physiological acceptor site and reduces the accumulation of PPIX. Here, we developed a specific strategy that uses transferrin receptor 1 (TRF1) as a Trojan horse to deliver V1 to erythroid progenitors. We designed a bifunctional peptide (P1-9R) including a TFR1-targeting peptide coupled to a nine-arginine cell-penetrating peptide (CPP) that facilitates the release of the AON from TFR1 in endosomal vesicles. We demonstrated that the P1-9R/V1 nanocomplex promotes the efficient and prolonged redirection of splicing towards the physiological splice site and subsequent normalization of WT FECH mRNA and protein levels. Finally, the P1-9R/V1 nanocomplex increases WT FECH mRNA production and significantly decreases PPIX accumulation in primary cultures of differentiating erythroid progenitors from an overt EPP-affected individual. P1-9R is a method designed to target erythroid progenitors and represents a potentially powerful tool for the in vivo delivery of therapeutic DNA in many erythroid disorders.


Assuntos
Antígenos CD/metabolismo , Peptídeos Penetradores de Células/metabolismo , Células Precursoras Eritroides/metabolismo , Terapia Genética/métodos , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/terapia , Receptores da Transferrina/metabolismo , Antígenos CD/administração & dosagem , Antígenos CD34/metabolismo , Linhagem Celular , Peptídeos Penetradores de Células/administração & dosagem , Eritroblastos/citologia , Eritroblastos/metabolismo , Ferroquelatase/genética , Ferroquelatase/metabolismo , Humanos , Ligantes , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Protoporfirinas/metabolismo , RNA Mensageiro , Receptores da Transferrina/administração & dosagem
8.
Vet Microbiol ; 219: 1-7, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29778179

RESUMO

Porcine reproductive and respiratory syndrome (PRRS) is one of the most important swine diseases affecting pig industry worldwide. Sialoadehesin (Sn) and CD163 are the two specific receptors for PRRSV infection of porcine alveolar macrophages. Our previous study showed that the soluble Sn receptor Sn4D-Fc and soluble CD163 receptor SRCR59-Fc expressed by the two recombinant adenoviral (rAd) vectors have an additive anti-PRRSV effect in vitro. In the present study, rAd-Sn4D-Fc and rAd-SRCR59-Fc were inoculated into pigs, and the efficient expression of Sn4D-Fc and SRCR59-Fc proteins was detected by ELISA. Then, PRRSV-naïve pigs were inoculated with rAd-Sn4D-Fc and/or rAd-SRCR59-Fc before contagious infection with different PRRSV strains. Among the three rAd inoculation groups, simultaneous inoculation with the two rAd vectors provided the best protection against highly pathogenic JXA1 strain PRRSV, followed by rAd-SRCR59-Fc inoculation and rAd-Sn4D-Fc inoculation. Clinical observation and quantitative RT-PCR analyses showed that all of the double rAd-inoculated pigs (n = 9) survived from the contagious infection with highly pathogenic JXA1, JS07 or SH1705 strain PRRSV with significantly alleviated clinical scores, viremia, fecal viral emission and tissue virus loads. These data suggest that rAd-Sn4D-Fc and rAd-SRCR59-Fc can be developed further as the universal therapeutic vaccine to facilitate PRRSV eradication.


Assuntos
Adenoviridae/genética , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Receptores de Superfície Celular/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Animais , Anticorpos Antivirais , Antígenos CD/administração & dosagem , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/administração & dosagem , Antígenos de Diferenciação Mielomonocítica/genética , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macrófagos Alveolares/virologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Receptores de Superfície Celular/administração & dosagem , Receptores de Superfície Celular/genética , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/administração & dosagem , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Suínos , Carga Viral , Viremia
9.
J Mol Neurosci ; 64(2): 200-210, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29280053

RESUMO

To explore the hypothesis that CD200Fc, a CD200R1 agonist with anti-inflammatory properties, will inhibit retinal glial cells hyperactivation and retinal ganglion cells (RGCs) apoptosis after optic nerve injury. CD200Fc was immediately administered after optic nerve crush (ONC) once by intravitreal injection. Rats were euthanized at 5 days after ONC. The density of RGCs was counted by immunostaining of retina flat mounts for Brn3a. TUNEL assay, immunoblotting analysis of ionized calcium-binding adapter molecule 1(iba1) (microglia marker) and glial fibrillary acidic protein (GFAP) (astrocytes and Müller cells marker), RT-PCR analysis of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin (IL)-8 and IL-10, ELISA measure protein levels of inflammatory cytokines and western blot analysis of CD200 and CD200R1 were evaluated. CD200Fc treatment suppressed ONC-induced RGCs loss through inhibition of RGCs apoptosis. Additionally, expression of glial cells activation markers GFAP and iba1 and production of pro-inflammatory cytokines (COX-2, iNOS, MCP-1, TNF-α, IL-8) were decreased in CD200Fc treated animals after ONC. Meanwhile, anti-inflammatory cytokine IL-10 was increased by CD200Fc treatment in ONC-induced rat retina. Finally, we found that CD200Fc significantly inhibited ONC-induced increased in expression of CD200 and raised the already high basal CD200R1 expression in the rat retina after ONC. Our results demonstrated that the anti-inflammatory effects of CD200Fc in ONC rats model through inhibited the activation of retinal glial cells via the interaction between CD200 and CD200R1, and the neuroprotective effects of CD200Fc on RGCs thought inhibited its apoptosis.


Assuntos
Antígenos CD/uso terapêutico , Apoptose , Neuroglia/efeitos dos fármacos , Traumatismos do Nervo Óptico/tratamento farmacológico , Receptores Imunológicos/metabolismo , Animais , Antígenos CD/administração & dosagem , Antígenos CD/farmacologia , Quimiocina CCL2/metabolismo , Ciclo-Oxigenase 2/metabolismo , Interleucina-8/metabolismo , Injeções Intravítreas , Masculino , Neuroglia/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ligação Proteica , Proteínas RGS/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Imunológicos/agonistas , Retina/citologia , Retina/efeitos dos fármacos , Retina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Int J Nanomedicine ; 12: 5557-5569, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28831252

RESUMO

Various strategies have been utilized to improve both gene transfer efficiency and cell-induced toxicity of polyethylenimine (PEI), the most extensively investigated cationic polymeric vector. In this study, we sought to enhance transfection efficiency of low molecular weight PEI (LMW PEI) while maintaining its low toxicity by cross-linking LMW PEI via succinic acid linker. These modifications were designed to improve the hydrophilic-hydrophobic balance of the polymer, by enhancing the buffering capacity and maintaining low cytotoxic effects of the final conjugate. Decreased expression of CD200 in the central nervous system has been considered as one of the proposed mechanisms associated with neuroinflammation in multiple sclerosis; therefore, we selected plasmid-encoding CD200 gene for transfection using the modified PEI derivatives. Dynamic light scattering experiments demonstrated that the modified PEIs were able to condense plasmid DNA and form polyplexes with a size of approximately 130 nm. The highest level of CD200 expression was achieved at a carrier to plasmid ratio of 8, where the expression level was increased by 1.5 fold in the SH-SY5Y cell line, an in vitro model of neurodegenerative disorders. Furthermore, the results of in vivo imaging of the LMW PEI-based nanoparticles in the mouse model of multiple sclerosis revealed that fluorescently labeled plasmid encoding CD200 was distributed from the injection site to various tissues and organs including lymph nodes, liver, brain, and finally, kidneys. The nanoparticles also showed the ability to cross the blood-brain barrier and enter the periventricular area.


Assuntos
Antígenos CD/genética , Nanopartículas/química , Polietilenoimina/química , Transfecção/métodos , Animais , Antígenos CD/administração & dosagem , Antígenos CD/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Difusão Dinâmica da Luz , Regulação da Expressão Gênica , Terapia Genética/métodos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Masculino , Camundongos Endogâmicos C57BL , Peso Molecular , Doenças Neurodegenerativas/terapia , Plasmídeos/administração & dosagem , Plasmídeos/genética , Distribuição Tecidual , Transgenes
11.
Biol Pharm Bull ; 39(8): 1309-18, 2016 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-27251665

RESUMO

An adequate immune response to percutaneous vaccine application is generated by delivery of sufficient amounts of antigen to skin and by administration of toxin adjuvants or invasive skin abrasion that leads to an adjuvant effect. Microneedles penetrate the stratum corneum, the outermost layer of the skin, and enable direct delivery of vaccines from the surface into the skin, where immunocompetent dendritic cells are densely distributed. However, whether the application of microneedles to the skin activates antigen-presenting cells (APCs) has not been demonstrated. Here we aimed to demonstrate that microneedles may act as a potent physical adjuvant for successful transcutaneous immunization (TCI). We prepared samples of isolated epidermal and dermal cells and analyzed the expression of major histocompatibility complex (MHC) class II and costimulatory molecules on Langerhans or dermal dendritic cells in the prepared samples using flow cytometry. The expression of MHC class II and costimulatory molecules demonstrated an upward trend in APCs in the skin after the application of 500- and 300-µm microneedles. In addition, in the epidermal cells, application of microneedles induced more effective activation of Langerhans cells than did an invasive tape-stripping (positive control). In conclusion, the use of microneedles is likely to have a positive effect not only as an antigen delivery system but also as a physical technique inducing an adjuvant-like effect for TCI.


Assuntos
Células Dendríticas/imunologia , Células de Langerhans/imunologia , Microinjeções , Agulhas , Pele/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígenos CD/administração & dosagem , Antígenos de Histocompatibilidade Classe II/administração & dosagem , Imunização , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pele/citologia
12.
J Control Release ; 232: 75-82, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27094605

RESUMO

The delivery of vaccines to the sublingual mucosa is an attractive prospect due to the ease and acceptability of such an approach. However, novel adjuvant and delivery approaches are required to optimally vaccinate at this site. We have previously shown that conjugation of protein antigen to the iron transport molecule, transferrin, can significantly enhance mucosal immune responses. We tested whether conjugating influenza haemagglutinin to transferrin could improve the immune response to sublingually delivered antigen. Transferrin conjugated haemagglutinin induced a significant antibody and T cell response in both naïve animals and previously immunized animals. The immune response generated was able to protect mice against influenza virus challenge. Sublingually administered antigen dispersed more widely through the gastro-intestinal tract than intranasally delivered antigen and transferrin conjugation had a more marked effect on sublingually delivered antigen than intranasal immunisation. From these studies we conclude that transferrin conjugation of antigen is effective at boosting immune responses to sublingually delivered antigen and may be an attractive approach for influenza vaccines, particularly when mass campaigns are required.


Assuntos
Antígenos CD/administração & dosagem , Hemaglutininas Virais/administração & dosagem , Vírus da Influenza A Subtipo H3N2 , Vacinas contra Influenza/administração & dosagem , Infecções por Orthomyxoviridae/prevenção & controle , Receptores da Transferrina/administração & dosagem , Administração Intranasal , Administração Sublingual , Animais , Anticorpos Antivirais/imunologia , Antígenos CD/química , Feminino , Hemaglutininas Virais/química , Imunoglobulina G/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/química , Pulmão/virologia , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Receptores da Transferrina/química , Linfócitos T/imunologia
13.
Cancer Sci ; 107(4): 398-406, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27079438

RESUMO

Therapeutic cancer vaccines are designed to treat cancer by boosting the endogenous immune system to fight against the cancer. In the development of clinically effective cancer vaccines, one of the most practical objectives is to identify adjuvants that are capable of optimizing the vaccine effects. In this study, we explored the potential of polyinosinic-polycytidylic acid (poly(I:C)) and LAG-3-Ig (soluble recombinant protein of lymphocyte activation gene-3 [LAG-3] extracellular domain fused with human IgG Fc region) as adjuvants for P1A tumor antigen peptide vaccine in a pre-established P815 mouse tumor model with a transfer of tumor-specific T cells. Whereas the use of poly(I:C) or LAG-3-Ig as a signal adjuvant induced a slight enhancement of P1A vaccine effects compared to incomplete Freund's adjuvant, combined treatment with poly(I:C) plus LAG-3-Ig remarkably potentiated antitumor effects, leading to complete rejection of pre-established tumor and long-term survival of mice. The potent adjuvant effects of poly(I:C) plus LAG-3-Ig were associated with an enhanced infiltration of T cells in the tumor tissues, and an increased proliferation and Th1-type cytokine production of tumor-reactive T cells. Importantly, the combined adjuvant of poly(I:C) plus LAG-3-Ig downregulated expressions of PD-1, LAG-3, and TIGIT on P1A-specific T cells, indicating prevention of T cell exhaustion. Taken together, the results of the current study show that the combined adjuvants of poly(I:C) plus LAG-3-Ig with tumor peptide vaccine induce profound antitumor effects by activating tumor-specific T cells.


Assuntos
Antígenos CD/imunologia , Vacinas Anticâncer/imunologia , Imunoglobulina G/imunologia , Poli I-C/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígenos CD/administração & dosagem , Antígenos CD/genética , Linfócitos T CD8-Positivos/efeitos dos fármacos , Humanos , Imunoglobulina G/administração & dosagem , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Poli I-C/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
14.
Oncogene ; 35(39): 5170-8, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27065336

RESUMO

Understanding what drives breast tumor progression is of utmost importance for blocking tumor metastasis; we have identified that semaphorin 7a is a potent driver of ductal carcinoma in situ (DCIS) progression. Semaphorin 7a is a glycophosphatidylinositol membrane-anchored protein that promotes attachment and spreading in multiple cell types. Here, we show that increased expression of SEMA7A occurs in a large percentage of breast cancers and is associated with decreased overall and distant metastasis-free survival. In both in vitro and in vivo models, short hairpin-mediated silencing of SEMA7A reveals roles for semaphorin 7a in the promotion of DCIS growth, motility and invasion as well as lymphangiogenesis in the tumor microenvironment. Our studies also uncover a relationship between COX-2 and semaphorin 7a expression and suggest that semaphorin 7a promotes tumor cell invasion on collagen and lymphangiogenesis via activation of ß1-integrin receptor. Our results suggest that semaphorin 7a may be novel target for blocking breast tumor progression.


Assuntos
Antígenos CD/genética , Neoplasias da Mama/genética , Carcinoma Intraductal não Infiltrante/genética , Ciclo-Oxigenase 2/genética , Neoplasias Mamárias Animais/genética , Semaforinas/genética , Animais , Antígenos CD/administração & dosagem , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/patologia , Movimento Celular/genética , Ciclo-Oxigenase 2/biossíntese , Progressão da Doença , Feminino , Proteínas Ligadas por GPI/administração & dosagem , Proteínas Ligadas por GPI/genética , Regulação Neoplásica da Expressão Gênica , Pleiotropia Genética , Humanos , Linfangiogênese/genética , Neoplasias Mamárias Animais/patologia , Camundongos , Semaforinas/administração & dosagem , Transdução de Sinais
15.
Expert Opin Ther Targets ; 20(7): 885-901, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26732941

RESUMO

INTRODUCTION: Semaphorin 4D (Sema4D), also known as CD100, has been implicated in physiologic roles in the immune and nervous systems. However, the interaction of Sema4D with its high affinity receptor, Plexin-B1, reveals a novel role for Sema4D produced by the tumor microenvironment in tumor angiogenesis and metastasis. AREAS COVERED: The ligation of Sema4D/CD100 with CD72 on immune and inflammatory cells is known to stimulate immune responses and regulation. Because CD100 and CD72 are expressed on lung immune and nonimmune cells, as well as on mast cells, the CD100/CD72 interaction plays another important role in allergic airway inflammation and mast cell functions. A better understanding of Sema4D-mediated cell signaling in physiological and pathological processes may be crucial for crafting new Sema4D-based therapeutics for human disease and tumor microenvironments. Strategies to achieve effective management through treatment with Sema4D include special siRNAs, neutralizing antibodies and knockdown. EXPERT OPINION: This review focuses on the links between Sema4D and human diseases such as cancer, bone metabolism, immune responses and organ development. The current knowledge regarding the expression of Sema4D and its receptors and its functional roles is systemically reviewed to explore Sema4D as both a target and a therapeutic in human diseases.


Assuntos
Antígenos CD/imunologia , Doenças Autoimunes/terapia , Terapia de Alvo Molecular , Semaforinas/imunologia , Animais , Antígenos CD/administração & dosagem , Doenças Autoimunes/imunologia , Doenças Ósseas/imunologia , Doenças Ósseas/terapia , Técnicas de Silenciamento de Genes , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Neovascularização Patológica/imunologia , Neovascularização Patológica/terapia , Neuroimunomodulação/imunologia , Semaforinas/administração & dosagem , Microambiente Tumoral
16.
Eur J Immunol ; 46(3): 689-700, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26678013

RESUMO

The development of new immunization strategies requires a better understanding of early molecular and cellular events occurring at the site of injection. The skin is particularly rich in immune cells and represents an attractive site for vaccine administration. Here, we specifically targeted vaccine antigens to epidermal Langerhans cells (LCs) using a fusion protein composed of HIV antigens and a monoclonal antibody targeting Langerin. We developed a fluorescence imaging approach to visualize, in vivo, the vaccine-targeted cells. Studies were performed in nonhuman primates (NHPs) because of their relevance as a model to assess human vaccines. We directly demonstrated that in NHPs, intradermally injected anti-Langerin-HIVGag specifically targets epidermal LCs and induces rapid changes in the LC network, including LC activation and migration out of the epidermis. Vaccine targeting of LCs significantly improved anti-HIV immune response without requirement of an adjuvant. Although the co-injection of the TLR-7/8 synthetic ligand, R-848 (resiquimod), with the vaccine, did not enhance significantly the antibody response, it stimulated recruitment of HLA-DR+ inflammatory cells to the site of immunization. This study allowed us to characterize the dynamics of early local events following the injection of a vaccine-targeted epidermal LCs and R-848.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antígenos CD/imunologia , Células de Langerhans/imunologia , Lectinas Tipo C/imunologia , Lectinas de Ligação a Manose/imunologia , Vacinas/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Antígenos CD/administração & dosagem , Células Epidérmicas , Epiderme/imunologia , Proteína do Núcleo p24 do HIV/administração & dosagem , Proteína do Núcleo p24 do HIV/imunologia , Humanos , Imidazóis/administração & dosagem , Imidazóis/imunologia , Injeções Intradérmicas , Microscopia Intravital , Células de Langerhans/ultraestrutura , Lectinas Tipo C/administração & dosagem , Macaca fascicularis , Lectinas de Ligação a Manose/administração & dosagem , Imagem Óptica , Vacinas/imunologia
17.
Hum Gene Ther ; 26(10): 647-56, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26076707

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene. It is the most common, severe childhood form of muscular dystrophy. We investigated an alternative to dystrophin replacement by overexpressing ITGA7 using adeno-associated virus (AAV) delivery. ITGA7 is a laminin receptor in skeletal muscle that, like the dystrophin-glycoprotein complex, links the extracellular matrix to the internal actin cytoskeleton. ITGA7 is expressed in DMD patients and overexpression does not elicit an immune response to the transgene. We delivered rAAVrh.74.MCK.ITGA7 systemically at 5-7 days of age to the mdx/utrn(-/-) mouse deficient for dystrophin and utrophin, a severe mouse model of DMD. At 8 weeks postinjection, widespread expression of ITGA7 was observed at the sarcolemma of multiple muscle groups following gene transfer. The increased expression of ITGA7 significantly extended longevity and reduced common features of the mdx/utrn(-/-) mouse, including kyphosis. Overexpression of α7 expression protected against loss of force following contraction-induced damage and increased specific force in the diaphragm and EDL muscles 8 weeks after gene transfer. Taken together, these results further support the use of α7 integrin as a potential therapy for DMD.


Assuntos
Antígenos CD/genética , Distrofina/genética , Cadeias alfa de Integrinas/genética , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animais , Antígenos CD/administração & dosagem , Antígenos CD/biossíntese , Dependovirus , Modelos Animais de Doenças , Distrofina/deficiência , Regulação da Expressão Gênica , Terapia Genética/métodos , Humanos , Cadeias alfa de Integrinas/administração & dosagem , Cadeias alfa de Integrinas/biossíntese , Camundongos , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia
18.
Oncol Rep ; 33(6): 2837-44, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25872647

RESUMO

The present study aimed to determine the effect of recombinant Salmonella (SL3261)-based CEACAM6 and 4-1BB ligand (4-1BBL) vaccine on the development of colorectal cancer in rats and the potential immune mechanisms involved. Attenuated Salmonella typhimurium (vaccine strain)­carrying plasmids pIRES-CEACAM6, pIRES­4­1BBL and pIRES-CEACAM6-4-1BBL were constructed. The rats were administered subcutaneous injections of 1,2-dimethyl-hydrazine (DMH) once a week for 18 weeks. Eight weeks after the first injection, the rats were divided into the pIRES/SL3261, pIRES-4-1BBL/SL3261, pIRES-CEACAM6/SL3261 and pIRES-CEACAM6-4-1BBL/SL3261 groups, and fed with corresponding vaccine strains. The rats were then sacrificed, the number of colon tumors were recorded, and the Dukes' stage were evaluated. CD3, CD4, CD8, CD56, FOXP3 and CEACAM6 expression in tumor tissues was determined by immunohistochemical staining. Compared with the expression levels in the pIRES/SL3261 group, similar levels of CD3+, CD8+ and CD56+ expression were identified for the pIRES-CEACAM6/SL3261 group of rats. Additionally, a comparable number of tumors was detected in the pIRES-4-1BBL/SL3261 and pIRES-CEACAM6/SL3261 groups. By contrast, a significantly fewer number of tumors, albeit with a higher density of CD3+CD8+, CD56+ and a lower density of Foxp3+ tumor-infiltrating lymphocyte (TIL) cells was detected in the pIRES-CEACAM6-4-1BBL/SL3261 group of rats. The results indicated that vaccination with recombinant attenuated Salmonella harboring the CEACAM6 and 4-1BBL gene efficiently increased the number of CD3+CD8+ TIL and NK cells, decreased the number of FOXP3 cells and inhibited the development of DMH-induced colorectal cancer in rats.


Assuntos
Ligante 4-1BB/imunologia , Antígenos CD/imunologia , Vacinas Anticâncer/imunologia , Moléculas de Adesão Celular/imunologia , Neoplasias Colorretais/imunologia , 1,2-Dimetilidrazina/toxicidade , Ligante 4-1BB/administração & dosagem , Ligante 4-1BB/genética , Animais , Antígenos CD/administração & dosagem , Antígenos CD/genética , Vacinas Anticâncer/administração & dosagem , Moléculas de Adesão Celular/administração & dosagem , Moléculas de Adesão Celular/genética , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/genética , Neoplasias Colorretais/terapia , Proteínas Ligadas por GPI/administração & dosagem , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Humanos , Imunoterapia , Linfócitos do Interstício Tumoral/imunologia , Ratos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Vacinas Atenuadas/imunologia
19.
Nat Commun ; 6: 6385, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25721933

RESUMO

Reproductive competence in mammals depends on the projection of gonadotropin-releasing hormone (GnRH) neurons to the hypothalamic median eminence (ME) and the timely release of GnRH into the hypothalamic-pituitary-gonadal axis. In adult rodents, GnRH neurons and the specialized glial cells named tanycytes periodically undergo cytoskeletal plasticity. However, the mechanisms that regulate this plasticity are still largely unknown. We demonstrate that Semaphorin7A, expressed by tanycytes, plays a dual role, inducing the retraction of GnRH terminals and promoting their ensheathment by tanycytic end feet via the receptors PlexinC1 and Itgb1, respectively. Moreover, Semaphorin7A expression is regulated during the oestrous cycle by the fluctuating levels of gonadal steroids. Genetic invalidation of Semaphorin7A receptors in mice induces neuronal and glial rearrangements in the ME and abolishes normal oestrous cyclicity and fertility. These results show a role for Semaphorin7A signalling in mediating periodic neuroglial remodelling in the adult ME during the ovarian cycle.


Assuntos
Antígenos CD/farmacologia , Eminência Mediana/fisiologia , Neuroglia/metabolismo , Plasticidade Neuronal/fisiologia , Semaforinas/farmacologia , Análise de Variância , Animais , Antígenos CD/administração & dosagem , Western Blotting , Ensaio de Imunoadsorção Enzimática , Estradiol/análogos & derivados , Feminino , Citometria de Fluxo , Imunofluorescência , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Ovariectomia , Progesterona , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Semaforinas/administração & dosagem
20.
G Ital Cardiol (Rome) ; 15(6): 355-62, 2014 Jun.
Artigo em Italiano | MEDLINE | ID: mdl-25072421

RESUMO

Recent advances in coronary revascularization techniques have improved the outcomes of ischemic heart disease in both acute and chronic settings. As a drawback, an increase in patients with an advanced stage of ischemic cardiomyopathy refractory to optimal medical treatment has been observed. Among the therapeutic alternatives under investigation, cell therapy showed considerable anti-ischemic potential. Although several types of cells have been used, bone marrow-derived endothelial progenitor cells are among the most appealing therapeutic agents due to their angiogenic properties. In particular, endothelial progenitors expressing the transmembrane protein CD133 have been in vitro and in vivo extensively characterized and clinically tested. The aim of this paper is to discuss the translational process that allowed the clinical application of CD133+ endothelial progenitor cells in the context of ischemic cardiomyopathy.


Assuntos
Antígenos CD/administração & dosagem , Células da Medula Óssea/citologia , Glicoproteínas/administração & dosagem , Injeções Intralesionais , Isquemia Miocárdica/terapia , Peptídeos/administração & dosagem , Transplante de Células-Tronco , Antígeno AC133 , Células da Medula Óssea/metabolismo , Ensaios Clínicos como Assunto , Medicina Baseada em Evidências , Humanos , Isquemia Miocárdica/metabolismo , Transplante de Células-Tronco/métodos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...