Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Oral Pathol Med ; 42(1): 9-16, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22563860

RESUMO

Photodynamic therapy (PDT) of cells is a new treatment modality involving selective delivery of a photosensitive dye into target cells, followed by visible light irradiation. PDT induces cell death by excessive ROS generation. The effects of multiple photosensitizers were owing to the difference in cell types involving sensitizer-specific protein changes linked to resistance. HSP27 is regulated in response to stress and is associated with apoptotic process. The effects of HSP27 on PDT resistance are controversial and unclear. The purpose of this study was to investigate the role of HSP27 down-regulation in the PDT-induced cells and HSP27 regulation in the resistance to PDT. KB cells transfected with HSP27 siRNA were exposed to hematoporphyrin (HP) followed by irradiation at 635 nm at an energy density of 4.5 mW/cm(2). After irradiation, the effects on HSP27 down-regulation were assessed by MTT assay, flow cytometry, confocal analysis, Western blotting and caspase activity. The results of this study showed that down-regulation of HSP27 restored cell survival in HP-PDT-induced apoptotic KB cells. HSP27 down-regulation attenuated PDT-induced apoptosis through caspase-mediated pathway in KB cells. Also, HSP27 silencing regulated Bax, Bcl-2, and PARP protein expression in PDT-induced cells. Therefore, HSP27 down-regulation confers resistance to PDT through interruption of apoptotic protein activity in PDT-induced cell death. HSP27 might contribute to regulating PDT-induced apoptosis in PDT-resistant cells.


Assuntos
Apoptose/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP27 , Neoplasias Bucais/tratamento farmacológico , Fotoquimioterapia , Caspases/genética , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo , Inativação Gênica , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/fisiologia , Humanos , Células KB , Neoplasias Bucais/enzimologia , Neoplasias Bucais/genética , RNA Interferente Pequeno
2.
J Immunother Cancer ; 11(4)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37019470

RESUMO

BACKGROUND: The incorporation of co-stimulatory signaling domains into second-generation chimeric antigen receptors (CARs) significantly enhances the proliferation and persistence of CAR-T cells in vivo, leading to successful clinical outcomes. METHODS: To achieve such functional enhancement in transgenic T-cell receptor-engineered T-cell (TCR-T) therapy, we designed a second-generation TCR-T cell in which CD3ζ genes modified to contain the intracellular domain (ICD) of the 4-1BB receptor were selectively inserted into the CD247 locus. RESULTS: This modification enabled the simultaneous recruitment of key adaptor molecules for signals 1 and 2 on TCR engagement. However, the addition of full-length 4-1BB ICD unexpectedly impaired the expression and signaling of TCRs, leading to suboptimal antitumor activity of the resulting TCR-T cells in vivo. We found that the basic-rich motif (BRM) in the 4-1BB ICD was responsible for the undesirable outcomes, and that fusion of minimal tumor necrosis factor receptor-associated factor (TRAF)-binding motifs at the C-terminus of CD3ζ (zBBΔBRM) was sufficient to recruit TRAF2, the key adaptor molecule in 4-1BB signaling, while retaining the expression and proximal signaling of the transgenic TCR. Consequently, TCR-T cells expressing zBBΔBRM exhibited improved persistence and expansion in vitro and in vivo, resulting in superior antitumor activity in a mouse xenograft model. CONCLUSIONS: Our findings offer a promising strategy for improving the intracellular signaling of TCR-T cells and their application in treating solid tumors.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Animais , Humanos , Camundongos , Modelos Animais de Doenças , Neoplasias/tratamento farmacológico , Receptores de Antígenos de Linfócitos T , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto , Sítios de Ligação
3.
Nat Med ; 28(9): 1802-1812, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35927581

RESUMO

Clearing amyloid-ß (Aß) through immunotherapy is one of the most promising therapeutic approaches to Alzheimer's disease (AD). Although several monoclonal antibodies against Aß have been shown to substantially reduce Aß burden in patients with AD, their effects on improving cognitive function remain marginal. In addition, a significant portion of patients treated with Aß-targeting antibodies experience brain edema and microhemorrhage associated with antibody-mediated Fc receptor activation in the brain. Here, we develop a phagocytosis inducer for Aß consisting of a single-chain variable fragment of an Aß-targeting monoclonal antibody fused with a truncated receptor binding domain of growth arrest-specific 6 (Gas6), a bridging molecule for the clearance of dead cells via TAM (TYRO3, AXL, and MERTK) receptors. This chimeric fusion protein (αAß-Gas6) selectively eliminates Aß plaques through TAM receptor-dependent phagocytosis without inducing NF-kB-mediated inflammatory responses or reactive gliosis. Furthermore, αAß-Gas6 can induce synergistic clearance of Aß by activating both microglial and astrocytic phagocytosis, resulting in better behavioral outcomes with substantially reduced synapse elimination and microhemorrhage in AD and cerebral amyloid angiopathy model mice compared with Aß antibody treatment. Our results suggest that αAß-Gas6 could be a novel immunotherapeutic agent for AD that overcomes the side effects of conventional antibody therapy.


Assuntos
Doença de Alzheimer , Anticorpos de Cadeia Única , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Animais , Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , NF-kappa B , Placa Amiloide/tratamento farmacológico , Receptores Fc/uso terapêutico , Anticorpos de Cadeia Única/uso terapêutico , c-Mer Tirosina Quinase
4.
World J Gastroenterol ; 10(8): 1191-7, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15069724

RESUMO

AIM: rAAV mediated endostatin gene therapy has been examined as a new method for treating cancer. However, a sustained and high protein delivery is required to achieve the desired therapeutic effects. We evaluated the impact of topoisomerase inhibitors in rAAV delivered endostatin gene therapy in a liver tumor model. METHODS: rAAV containing endostatin expression cassettes were transduced into hepatoma cell lines. To test whether the topoisomerase inhibitor pretreatment increased the expression of endostatin, Western blotting and ELISA were performed. The biologic activity of endostatin was confirmed by endothelial cell proliferation and tube formation assays. The anti-tumor effects of the rAAV-endostatin vector combined with a topoisomerase inhibitor, etoposide, were evaluated in a mouse liver tumor model. RESULTS: Topoisomerase inhibitors, including camptothecin and etoposide, were found to increase the endostatin expression level in vitro. The over-expressed endostatin, as a result of pretreatment with a topoisomerase inhibitor, was also biologically active. In animal experiments, the combined therapy of topoisomerase inhibitor, etoposide with the rAAV-endostatin vector had the best tumor-suppressive effect and tumor foci were barely observed in livers of the treated mice. Pretreatment with an etoposide increased the level of endostatin in the liver and serum of rAAV-endostatin treated mice. Finally, the mice treated with rAAV-endostatin in combination with etoposide showed the longest survival among the experimental models. CONCLUSION: rAAV delivered endostatin gene therapy in combination with a topoisomerase inhibitor pretreatment is an effective modality for anticancer gene therapy.


Assuntos
Adenoviridae/genética , Camptotecina/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Terapia Genética/métodos , Neoplasias Hepáticas/tratamento farmacológico , Sarcoma/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Carcinoma Hepatocelular/mortalidade , Linhagem Celular Tumoral , Terapia Combinada , Sinergismo Farmacológico , Endostatinas/farmacologia , Endotélio Vascular/citologia , Etoposídeo/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/mortalidade , Camundongos , Sarcoma/mortalidade , Taxa de Sobrevida , Inibidores da Topoisomerase , Veias Umbilicais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA