Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 12(1)2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-31936895

RESUMO

Targeted therapy is an efficient treatment for patients with epidermal growth factor receptor (EGFR) mutations in non-small cell lung cancer (NSCLC). Therapeutic resistance invariably occurs in NSCLC patients. Many studies have focused on drug resistance mechanisms, but only a few have addressed the metabolic flexibility in drug-resistant NSCLC. In the present study, we found that during the developing resistance to tyrosine kinase inhibitor (TKI), TKI-resistant NSCLC cells acquired metabolic flexibility in that they switched from dependence on glycolysis to oxidative phosphorylation by substantially increasing the activity of the mitochondria. Concurrently, we found the predominant expression of monocarboxylate transporter 1 (MCT-1) in the TKI-resistant NSCLC cells was strongly increased in those cells that oxidized lactate. Thus, we hypothesized that inhibiting MCT-1 could represent a novel treatment strategy. We treated cells with the MCT-1 inhibitor AZD3965. We found a significant decrease in cell proliferation and cell motility in TKI-sensitive and TKI-resistant cells. Taken together, these results demonstrated that gefitinib-resistant NSCLC cells harbored higher mitochondrial bioenergetics and MCT-1 expression. These results implied that targeting mitochondrial oxidative phosphorylation proteins or MCT-1 could serve as potential treatments for both TKI-sensitive and -resistant non-small cell lung cancer.

2.
Oncotarget ; 6(33): 35073-86, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26551021

RESUMO

Our previous studies suggest that antibodies against ENO1 (anti-ENO1 Ab) have a protective role in patients with non-small cell lung carcinoma. In this study, we evaluated the prognostic value of anti-ENO1 Ab levels in non-small cell lung carcinoma patients undergoing surgery. Circulating levels of anti-ENO1 Ab were assessed in 85 non-small cell lung carcinoma patients before and after surgery, and were correlated with clinical outcome. After surgery, patients with a higher increase of anti-ENO1 Ab had a lower hazard ratio and a better progression-free survival. Using animal models, we demonstrated that tumor cells reduce the circulating levels of anti-ENO1 Ab through physical absorption and neutralization of anti-ENO1 Ab with surface-expressed and secreted ENO1, respectively. Mice transplanted with ENO1-overexpressing tumors generated ENO1-specific regulatory T cells to suppress the production of anti-ENO1 Ab. Our results suggest that the increase of anti-ENO1 Ab may reflect anti-tumor immune responses and serve as a prognostic marker in postoperative lung cancer patients.


Assuntos
Anticorpos Antineoplásicos/sangue , Biomarcadores Tumorais/imunologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Proteínas de Ligação a DNA/imunologia , Neoplasias Pulmonares/imunologia , Fosfopiruvato Hidratase/imunologia , Proteínas Supressoras de Tumor/imunologia , Idoso , Animais , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/sangue , Carcinoma Pulmonar de Células não Pequenas/sangue , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Intervalo Livre de Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/mortalidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Prognóstico
3.
PLoS One ; 8(7): e69354, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894455

RESUMO

In previous research, we found α-enolase to be inversely correlated with progression-free and overall survival in lung cancer patients and detected α-enolase on the surface of lung cancer cells. Based on these findings, we hypothesized that surface α-enolase has a significant role in cancer metastasis and tested this hypothesis in the current study. We found that α-enolase was co-immunoprecipitated with urokinase-type plasminogen activator, urokinase-type plasminogen activator receptor, and plasminogen in lung cancer cells and interacted with these proteins in a cell-free dot blotting assay, which can be interrupted by α-enolase-specific antibody. α-Enolase in lung cancer cells co-localized with these proteins and was present at the site of pericellular degradation of extracellular matrix components. Treatment with antibody against α-enolase in vitro suppressed cell-associated plasminogen and matrix metalloproteinase activation, collagen and gelatin degradation, and cell invasion. Examination of the effect of treatment with shRNA plasmids revealed that down regulation of α-enolase decreases extracellular matrix degradation by and the invasion capacity of lung cancer cells. Adoptive transfer of α-enolase-specific antibody to mice resulted in accumulation of antibody in subcutaneous tumor and inhibited the formation of tumor metastasis in lung and bone. This study demonstrated that surface α-enolase promotes extracellular matrix degradation and invasion of cancer cells and that targeting surface α-enolase is a promising approach to suppress tumor metastasis.


Assuntos
Membrana Celular/metabolismo , Matriz Extracelular/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Fosfopiruvato Hidratase/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacologia , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/metabolismo , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Movimento Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Humanos , Hospedeiro Imunocomprometido , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Metástase Neoplásica , Neoplasias/mortalidade , Fosfopiruvato Hidratase/antagonistas & inibidores , Plasminogênio/metabolismo , Ligação Proteica , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Immunother ; 32(4): 363-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19342969

RESUMO

Macrophage inflammation protein-3alpha (MIP-3alpha) is a chemokine expressed in inflamed tissue and capable of inducing migration of immature dendritic cells (DCs) or Langerhans cells. We postulated that conditioning vaccination sites with MIP-3alpha might enhance the efficacy of subsequently administered DC-based cancer vaccines. Our results demonstrate that subcutaneously injection of irradiated tumor cells expressing MIP-3alpha induces substantial cell infiltration to the injection site. Vaccination of irradiated tumor cells expressing MIP-3alpha followed by DCs pulsed with irradiated tumor cells can effectively suppress tumor growth in animals, which is significantly better than vaccination with irradiated MIP-3alpha-producing tumor cells or DCs pulsed with tumor cells alone. The protective effect was most evident when the MIP-3alpha-producing tumor cells and DC-based vaccines were injected at the same site. These results support the notion that this combination vaccination strategy might generate a more effective immune response to suppress the growth of tumor cells in animals.


Assuntos
Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Quimiocina CCL20/imunologia , Células Dendríticas/imunologia , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Movimento Celular , Células Dendríticas/transplante , Neoplasias Pulmonares/secundário , Melanoma Experimental/prevenção & controle , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Transfecção
5.
Cancer ; 103(4): 763-71, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15637694

RESUMO

BACKGROUND: The authors conducted a pilot clinical trial to explore the vaccination of patients with late-stage lung carcinoma with dendritic cells (DCs) pulsed with necrotic tumor cells derived from malignant pleural effusion specimens, and to evaluate the antitumor immune response induced by this therapy. METHODS: Autologous DCs were generated by culturing adherent mononuclear cells with interleukin-4 and granulocyte-macrophage-colony-stimulating factor for 7 days. Day-7 DCs were cocultured overnight with autologous necrotic tumor cells derived from pleural effusion specimens to allow internalization of tumor antigens. DCs were then treated with tumor necrosis factor-alpha for 16 hours. The antigen-loaded DCs were injected into each patient's inguinal lymph nodes under sonographic guidance. Eight patients with late-stage nonsmall cell lung carcinoma were treated in this manner. Patients were vaccinated once weekly for 4 weeks and then boosted twice biweekly. RESULTS: The authors found that there was no Grade II/III toxicity and autoimmune response in all patients after intranodal injection of the DC vaccine. Minor to moderate increases in T-cell responses against tumor antigens were observed after DC vaccination in six of eight patients. Five patients had progressive disease. One patient had minor tumor response and two patients had stable disease. The two patients who had longer disease control also had better T-cell responses. CONCLUSIONS: The results indicated that it was feasible to immunize patients with lung carcinoma intranodally with DCs pulsed with necrotic tumor cells enriched from pleural effusion specimens, and this approach may generate T-cell responses and provide clinical benefit in some patients.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/terapia , Células Dendríticas/transplante , Neoplasias Pulmonares/terapia , Derrame Pleural Maligno/citologia , Adulto , Idoso , Antígenos de Neoplasias/imunologia , Técnicas de Cocultura , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interleucina-4/farmacologia , Teste de Cultura Mista de Linfócitos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/farmacologia
6.
Biochem Pharmacol ; 68(2): 293-303, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15194001

RESUMO

BPR0Y007, a bis-benzylidenecyclopentanone derivative (2,5-bis- (4-hydroxy-3-methoxybenzylidene) cyclopentanone), was identified in our laboratory as a novel antineoplastic agent with a broad spectrum of antitumor activity against many human cancer cells. A previous study showed that BPR0Y007 inhibited DNA topoisomerase I (Top 1) activity and prevented tubulin polymerization. Notably, no cross-resistance with BPR0Y007 was observed in camptothecin-, VP-16- or vincristine-resistant cell lines. In this study, we further investigated the cellular and molecular events underlying the antitumoral function of this compound in human oral epidermoid carcinoma KB cells, focusing on the early cytotoxic effect. Treatment of KB cells with BPR0Y007-induced G(2)/M phase arrest followed by sub-G(1) phase accumulation. Annexin-V-propidium iodide (PI) binding assay and DNA fragmentation assay further indicated that BPR0Y007-induced cell death proceeded through an apoptotic pathway as opposed to via necrosis. This compound produced a time-dependent activation of caspases-3 and -8, however, another caspase-3 initiator, caspase-9, was only marginally activated at later time point. We further demonstrated that the activation of the caspases cascade and nuclear fragmentation was not associated with inactivated Bcl-2 and perturbed mitochondrial membrane potential by BPR0Y007. The finding that BPR0Y007-induced apoptosis through a membrane-mediated mechanism was supported by up-regulated expression of Fas (CD95/APO-1), but not Fas-L. Furthermore, up-regulation of p53 and its affected gene, MDM2, in KB cells was found after BPR0Y007 exposure. Overall, our results demonstrated that the BPR0Y007 could induce an early cytotoxic apoptosis through a caspase-8-dependent but mitochondrial-caspase-9 independent pathway, and involving upregulation of p53.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Compostos de Benzilideno/farmacologia , Caspases/metabolismo , Pentanonas/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Receptor fas/metabolismo , Carcinoma de Células Escamosas/patologia , Caspase 3 , Caspase 8 , Caspase 9 , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Proteína Ligante Fas , Fase G2/efeitos dos fármacos , Humanos , Células KB , Glicoproteínas de Membrana/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Mitose/efeitos dos fármacos , Proteínas/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos
7.
J Interferon Cytokine Res ; 24(4): 231-43, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15144569

RESUMO

Various human colon cancer cell lines tested in vitro differed significantly in susceptibility to growth inhibition of recombinant human interferon-beta (rHuIFN-beta). Two p53-mutant lines, COH and CC-M2, derived from high-grade colon adenocarcinoma, showed signs of apoptosis after treatment with 250 IU/ml of HuIFN- beta in the culture medium. The similarly p53-mutated HT-29 line from a grade I adenocarcinoma showed no apoptosis, however, and only cell cycle G1/G0 or S phase retardation with 1000 IU/ml HuIFN-beta. After HuIFN-beta exposure, COH and CC-M2 cells showed increased levels of Fas and FasL proteins, alteration of mitochondrial membrane potential, and activation of caspase-9, caspase-8, and caspase-3 in a time-dependent manner. Treatment of COH and CC-M2 cells with anti-FasL antibodies or rFas/Fc fusion protein, however, could not prevent the apoptosis induced by HuIFN-beta. In contrast, cell-permeable specific inhibitors of the three caspases could inhibit the DNA fragmentation and cell death but not the mitochondrial membrane potential changes. Treatment with mitochondria-stabilizing reagents could significantly abrogate the apoptosis and caspase activation induced by HuIFN-beta. These results suggest that in COH and CC-M2 colon cancer cell lines, HuIFN-beta induces apoptosis mainly through mitochondrial membrane alteration and subsequent activation of the caspase cascade pathway, but not by the Fas/FasL interaction or the p53-dependent apoptotic mechanism.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Interferon Tipo I/farmacologia , Mitocôndrias/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2 , Receptores do Fator de Necrose Tumoral , Adenocarcinoma/enzimologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Antineoplásicos/toxicidade , Inibidores de Caspase , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Citocromos c/metabolismo , Ativação Enzimática/efeitos dos fármacos , Proteína Ligante Fas , Genes p53/genética , Humanos , Interferon Tipo I/toxicidade , Glicoproteínas de Membrana/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mutação/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Recombinantes , Proteína X Associada a bcl-2 , Receptor fas
8.
Clin Cancer Res ; 10(8): 2645-51, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15102666

RESUMO

PURPOSE: We intranodally immunized metastatic colorectal carcinoma patients, who had failed standard chemotherapy, with dendritic cells (DCs) pulsed with HLA-A*0201- or HLA-A*2402-restricted carcinoembryonic antigen (CEA) peptides to evaluate the safety of this treatment and the immune response against CEA peptides before and after the treatment. EXPERIMENTAL DESIGN: Six patients with the HLA-A*2402 genotype and 4 patients with the HLA-A*0201 genotype were enrolled. A single CEA peptide (YLSGANLNL) or two CEA peptides (QYSWFVNGTF and TYACFVSNL) were used for patients with the HLA-A*0201 or HLA-A*2402 genotype, respectively. Autologous DCs were generated by culturing adherent mononuclear cells with interleukin 4 and granulocyte macrophage colony-stimulating factor for 6 days. Maturation of DCs was then induced with tumor necrosis factor alpha for 40 h. Mature DCs were pulsed with appropriate CEA peptides for 2 h. After washing, 1 million peptide-pulsed DCs were injected into one inguinal lymph node under sonographic guidance. Each patient received four injections. RESULTS: No grade II/III toxicity or autoimmunity was observed. An increase in the number of CEA-specific T cells after DC vaccination could be detected in 7 of 10 (70%) patients. Two (20%) patients had stable disease for at least 12 weeks. One of these 2 patients experienced a transient decrease in CEA levels during the treatment period and also had the most significant T-cell response against the immunizing CEA peptides. CONCLUSIONS: These results suggest that our vaccination procedure can generate or boost specific T-cell responses and may provide clinical benefit in certain cancer patients.


Assuntos
Vacinas Anticâncer , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/terapia , Células Dendríticas/metabolismo , Antígenos HLA-A/genética , Peptídeos/uso terapêutico , Linfócitos T/imunologia , Adulto , Idoso , Células Cultivadas , Neoplasias Colorretais/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Genótipo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Antígeno HLA-A2 , Antígeno HLA-A24 , Humanos , Imunoterapia/métodos , Interferon gama/metabolismo , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Peptídeos/química , Linfócitos T/metabolismo , Fatores de Tempo
9.
Pharm Res ; 21(1): 93-100, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14984262

RESUMO

PURPOSE: The G-quadruplex ligand 3,3'-diethyloxadicarbocyanine iodide (DODC) was reported to enhance the apoptotic potency of pheochromocytoma PC-12 and leukemia HL-60 cells through the inhibition of telomerase activity. In this study, a mitochondrion-mediated apoptotic pathway was demonstrated as another cytotoxic mechanism for DODC action. METHODS: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and DNA laddering assays were performed to exhibit the cytotoxicity and apoptosis-inducing activity of DODC. Telomeric repeat amplification protocol (TRAP) assay was used to evaluate the effect of DODC on cellular telomerase. The mitochondrial uptake of probe 3,3'-dihexyloxacarbocyanine iodide was measured by flow cytometry. The mitochondrial proteomes were analyzed by two-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS). Western blot analyses were adopted to demonstrate the change of the distribution of mitochondrial proteins. RESULTS: DODC alone was able to induce apoptotic cell death but not decrease of telomerase activity in nasopharyngeal carcinoma NPC-TW01 cells. Instead, we found evidence that DODC significantly affected cellular mitochondria. DODC inhibited the uptake of another mitochondrial probe 3,3'-dihexyloxacarbocyanine iodide. By proteomic comparative analysis, we found that DODC induced the increase of prohibitin level in the mitochondria, indicating the occurrence of mitochondrial perturbation. Moreover, DODC was found to induce the levels of p53 and an 18-kDa truncated Bax on mitochondria, which in turn potentiated the release of cytochrome c for activation of caspases. CONCLUSIONS: DODC induces NPC-TW01 cell apoptosis via a mitochondrion-mediated mechanism. This paper demonstrates another cytotoxic mechanism of DODC other than inhibition of telomerase.


Assuntos
Apoptose/fisiologia , Carbocianinas/metabolismo , DNA/metabolismo , Guanosina/metabolismo , Mitocôndrias/fisiologia , Neoplasias Nasofaríngeas/enzimologia , Neoplasias Nasofaríngeas/patologia , Telomerase/metabolismo , Apoptose/efeitos dos fármacos , Carbocianinas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Quadruplex G , Humanos , Ligantes , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Telomerase/antagonistas & inibidores
10.
Int J Oncol ; 24(3): 581-90, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14767543

RESUMO

The accumulated results of recent clinical studies have indicated that aberrant epidermal growth factor receptor (EGFR) activation due to gene amplification and/or rearrangement contributes to increased malignancy and poor prognosis in many human cancers, especially in human glioblastoma multiforme (GBM). The elevated EGFR signaling in GBM has been correlated with shorter interval to relapse and lower survival rates, even in patients treated with surgery, radiation therapy, and/or chemotherapy. Therefore, the blockade of EGFR signaling in GBM may provide an ideal alternative therapeutic strategy. In this study, two EGFR-overexpressing human GBM cell lines (i.e., DBTRG and GBM 8901) were used as a model system. We demonstrated that expression of a human EGFR (EGFRt-EGFP) chimera protein in which the cytoplasmic domain is substituted by EGFP significantly reduced the EGF-induced endogenous EGFR autophosphorylation, EGF-induced downstream extra-cellular signal-regulated kinase (ERK) and Akt signaling, and the proportion of internalized receptors in EGF stimulated cells. Furthermore, these cells' anchorage-independent growth in vitro was decreased and their tumorigenicity in vivo abrogated or strongly suppressed. Our data suggest that EGFRt-EGFP abrogates tumor growth by disrupting receptor activation via competing for EGF-like ligands, forming non-activated heterodimers with endogenous EGFR, and inhibiting the EGFR endosomal signaling by substantially diminishing receptor internalization. This treatment modality (termed 'dominant-negative EGFR therapy') and its efficacy for gliomas or other tumors are under scrutiny.


Assuntos
Neoplasias do Sistema Nervoso Central/metabolismo , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Animais , Biotinilação , Western Blotting , Linhagem Celular Tumoral , Neoplasias do Sistema Nervoso Central/terapia , Dimerização , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Genes Dominantes , Vetores Genéticos , Glioblastoma/terapia , Proteínas de Fluorescência Verde , Humanos , Ligantes , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Testes de Precipitina , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes/metabolismo , Sefarose/química , Transdução de Sinais , Fatores de Tempo
11.
Cancer Gene Ther ; 11(2): 135-47, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14647233

RESUMO

To improve the efficacy of tumor cell-based and dendritic cell (DC)-based cancer vaccines, this study explored the potential of a new cancer vaccine strategy, that is, the use of CD40 ligand-transfected tumor (CD40L-tumor) cells to simultaneously deliver both tumor-derived antigens (Ag) and maturation stimuli to DCs. Materials from frozen/thawed or irradiated human tumor cells, with or without surface CD40L, were internalized efficiently by immature DCs after coincubation. However, during the internalization process, only coculturing with irradiated CD40L-tumor cells resulted in concurrent, optimal DC maturation and production of proinflammatory chemokines and pro-Th1 cytokines, such as IL-6, IL-8, IL-12, IFN-gamma, and TNF-alpha. These activated DCs were the most potent cells to support the growth of CD8+, IFN-gamma-producing T cells, and to process tumor Ag for the generation of specific cytotoxic T cells in vitro. Animals vaccinated with irradiated CD40L-tumor cell-pulsed DCs were better protected against subsequent challenge of a weakly immunogenic tumor cell line than animals vaccinated with irradiated CD40L-tumor cells alone. Thus, our results strongly support the future clinical application of using DCs pulsed with irradiated CD40L-tumor cells as a cancer vaccine.


Assuntos
Antígenos de Neoplasias/imunologia , Ligante de CD40/genética , Vacinas Anticâncer , Células Dendríticas/imunologia , Neoplasias Experimentais/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos CD , Ligante de CD40/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/metabolismo , Células Dendríticas/transplante , Humanos , Imunoglobulinas/metabolismo , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Camundongos , Neoplasias Experimentais/imunologia , Células Th1/imunologia , Células Th2/imunologia , Transfecção , Antígeno CD83
12.
Am J Physiol Cell Physiol ; 285(3): C711-9, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12736135

RESUMO

Regulated expression systems are invaluable for studying gene function, offer advantages of dosage-dependent and temporally defined gene expression, and limit possible clonal variation when toxic or pleiotropic genes are overexpressed. Previously, establishment of inducible expression systems, such as tetracycline- and ecdysone-inducible systems, required assessment of the inducible characteristics of individual clones by tedious luciferase assays. Taking advantage of a green fluorescent protein (GFP) reporter controlled by tetracycline- or ecdysone-responsive element and fluorescence-activated cell sorting, we propose a simple and efficient strategy to select highly inducible cell lines according to their fluorescence profiles after transiently transfecting the candidate cell pools with a surrogate GFP reporter. We have demonstrated that tetracycline- and ecdysone-inducible systems could be set up in Madin-Darby canine kidney and HEK-293 cells by employing this selection scheme. Importantly, this dual regulatory expression system is applied in studying the complex interplay between two Ras-related small GTPases, Cdc42 and Rac1, on detachment-induced apoptosis. Furthermore, establishment of two tightly regulated expression systems in one target cell line could be of great advantage for dissecting small GTPase Rac1-transduced signaling pathways by using global gene expression approaches such as proteomic assays.


Assuntos
Ecdisona/farmacologia , Biologia Molecular/métodos , Tetraciclinas/farmacologia , Proteínas rac1 de Ligação ao GTP/genética , Animais , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes/metabolismo , Rim/citologia , Proteínas Luminescentes/genética , Proteômica , Transdução de Sinais/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...