Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Total Environ ; 912: 168949, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38042186

RESUMO

Di-2-ethylhexyl phthalic acid (DEHP) is one of the most widely used plasticizers in the industry, which can improve the flexibility and durability of plastics. It is prone to migrate from various daily plastic products through wear and leaching into the surrounding environment and decompose into the more toxic metabolite mono-2-ethylhexyl phthalic acid (MEHP) after entering the human body. However, the impacts and mechanisms of MEHP on neuroblastoma are unclear. We exposed MYCN-amplified neuroblastoma SK-N-BE(2)C cells to an environmentally related concentration of MEHP and found that MEHP increased the proliferation and migration ability of tumor cells. The peroxisome proliferator-activated receptor (PPAR) ß/δ pathway was identified as a pivotal signaling pathway in neuroblastoma, mediating the effects of MEHP through transcriptional sequencing analysis. Because MEHP can bind to the PPARß/δ protein and initiate the expression of the downstream gene angiopoietin-like 4 (ANGPTL4), the PPARß/δ-specific agonist GW501516 and antagonist GSK3787, the recombinant human ANGPTL4 protein, and the knockdown of gene expression confirmed the regulation of the PPARß/δ-ANGPTL4 axis on the malignant phenotype of neuroblastoma. Based on the critical role of PPARß/δ and ANGPTL4 in the metabolic process, a non-targeted metabolomics analysis revealed that MEHP altered multiple metabolic pathways, particularly lipid metabolites involving fatty acyls, glycerophospholipids, and sterol lipids, which may also be potential factors promoting tumor progression. We have demonstrated for the first time that MEHP can target binding to PPARß/δ and affect the progression of neuroblastoma by activating the PPARß/δ-ANGPTL4 axis. This mechanism confirms the health risks of plasticizers as tumor promoters and provides new data support for targeted prevention and treatment of neuroblastoma.


Assuntos
Dietilexilftalato/análogos & derivados , Neuroblastoma , PPAR delta , PPAR beta , Ácidos Ftálicos , Humanos , PPAR beta/agonistas , PPAR beta/genética , PPAR beta/metabolismo , Proteína Proto-Oncogênica N-Myc , Plastificantes/toxicidade , Angiopoietinas/genética , Angiopoietinas/metabolismo , Ácidos Ftálicos/toxicidade , Ácidos Ftálicos/metabolismo , PPAR delta/agonistas , PPAR delta/genética , PPAR delta/metabolismo , Proteína 4 Semelhante a Angiopoietina
2.
Cell Tissue Res ; 394(3): 455-469, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37907763

RESUMO

Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) contribute to the intimal hyperplasia in type 2 diabetes mellitus (T2DM) patients after percutaneous coronary intervention. We aimed to investigate the role of lncRNA cyclin-dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) in VSMC proliferation and migration, as well as the underlying mechanism. T2DM model mice with carotid balloon injury were used in vivo and mouse aortic vascular smooth muscle cells (MOVAS) stimulated by insulin were used in vitro to assess the role of CDKN2B-AS1 in VSMC proliferation and migration following vascular injury in T2DM state. To investigate cell viability and migration, MTT assay and Transwell assay were conducted. To elucidate the underlying molecular mechanisms, the methylation-specific polymerase chain reaction, RNA immunoprecipitation, RNA-pull down, co-immunoprecipitation, and chromatin immunoprecipitation were performed. In vivo, CDKN2B-AS1 was up-regulated in common carotid artery tissues. In vitro, insulin treatment increased CDKN2B-AS1 level, enhanced MOVAS cell proliferation and migration, while the promoting effect was reversed by CDKN2B-AS1 knockdown. CDKN2B-AS1 forms a complex with enhancer of zeste homolog 2 (EZH2) and DNA methyltransferase (cytosine-5) 1 (DNMT1) to regulate smooth muscle 22 alpha (SM22α) methylation levels. In insulin-stimulated cells, SM22α knockdown abrogated the inhibitory effect of CDKN2B-AS1 knockdown on cell viability and migration. Injection of lentivirus-sh-CDKN2B-AS1 relieved intimal hyperplasia in T2DM mice with carotid balloon injury. Up-regulation of CDKN2B-AS1 induced by insulin promotes cell proliferation and migration by targeting SM22α through forming a complex with EZH2 and DNMT1, thereby aggravating the intimal hyperplasia after vascular injury in T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , RNA Longo não Codificante , Lesões do Sistema Vascular , Humanos , Camundongos , Animais , Hiperplasia , Músculo Liso Vascular/patologia , Insulina/farmacologia , Lesões do Sistema Vascular/patologia , RNA Longo não Codificante/genética , Proliferação de Células , Movimento Celular , Miócitos de Músculo Liso/patologia
3.
Biol Chem ; 403(3): 317-329, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34882999

RESUMO

Abnormal proliferation of vascular smooth muscle cells (VSMCs) induced by insulin resistance facilitates intimal hyperplasia of type 2 diabetes mellitus (T2DM) and N6-methyladenosine (m6A) methylation modification mediates the VSMC proliferation. This study aimed to reveal the m6A methylation modification regulatory mechanism. In this study, m6A demethylase FTO was elevated in insulin-treated VSMCs and T2DM mice with intimal injury. Functionally, FTO knockdown elevated m6A methylation level and further restrained VSMC proliferation and migration induced by insulin. Mechanistically, FTO knockdown elevated Smooth muscle 22 alpha (SM22α) expression and m6A-binding protein IGF2BP2 enhanced SM22α mRNA stability by recognizing and binding to m6A methylation modified mRNA. In vivo studies confirmed that the elevated m6A modification level of SM22α mRNA mitigated intimal hyperplasia in T2DM mice. Conclusively, m6A methylation-mediated elevation of SM22α restrained VSMC proliferation and migration and ameliorated intimal hyperplasia in T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Insulinas , Animais , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Insulinas/metabolismo , Metilação , Camundongos , Músculo Liso Vascular/patologia , RNA Mensageiro/metabolismo
4.
Ann Palliat Med ; 10(2): 2072-2079, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33615816

RESUMO

BACKGROUND: To assess the prevalence and associated risk of potentially inappropriate medications (PIMs) prescribing in community-dwelling elderly patients in China and to examine the most frequently used PIMs. This will provide a reference for the formulation of medication manuals for the community-dwelling elderly and further standardize the use of medications in elderly patients. METHODS: We conducted a cross-sectional retrospective study from April 1, 2020 to April 30, 2020. Data from elderly patients aged ≥65 years were collected from the Hengjie (N=2,294), Loujiang (N=3,972), and Tongxing communities (N=1,969) in Suzhou. The frequency of PIMs was detected using the 2019 Beers criteria and the 2017 Chinese criteria. Chi-square (for categorical variables), ANOVA (for continuous variables as applicable), and logistic regression were used to describe and identify potential predictors of PIMs. RESULTS: A total of 8,235 elderly patients were examined. Using the Chinese criteria, the prevalence of PIMs was 37.07%, which was slightly higher than that found using the 2019 Beers criteria (32.16%). The most prescribed PIMs were estazolam (21.53%) and insulin (4.60%) based on the Chinese criteria. Logistic regression analysis showed that advanced age, polypharmacy, and comorbid disease of patients were associated with a high risk of PIMs. Furthermore, the educational background and professional title of physicians were also associated with PIMs. CONCLUSIONS: Given the high prevalence of PIMs in the Chinese community-dwelling elderly population, the implementation of evidence-based interventions to promote rational clinical drug use could improve their quality of life.


Assuntos
Lista de Medicamentos Potencialmente Inapropriados , Qualidade de Vida , Idoso , China , Estudos Transversais , Humanos , Prescrição Inadequada , Prevalência , Estudos Retrospectivos
5.
Mol Ther Nucleic Acids ; 20: 62-72, 2020 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-32146419

RESUMO

Inflammation and proliferation of vascular smooth muscle cells (VSMCs) are the key events in intimal hyperplasia. This study aimed to explore the mechanism by which long non-coding RNA (lncRNA) KCNQ1OT1 affects VSMC inflammation and proliferation in this context. A vein graft (VG) model was established in mice to introduce intimal hyperplasia. Isolated normal VSMCs were induced with platelet-derived growth factor type BB (PDGF-BB), and the cell proliferation, migration, and secretion of inflammatory factors were determined. The results showed that KCNQ1OT1 was downregulated in the VSMCs from mice with intimal hyperplasia and in the PDGF-BB-treated VSMCs, and such downregulation of KCNQ1OT1 resulted from the increased methylation level in the KCNQ1OT1 promoter. Overexpressing KCNQ1OT1 suppressed PDFG-BB-induced VSMC proliferation, migration, and secretion of inflammatory factors. In VSMCs, KCNQ1OT1 bound to the nuclear transcription factor kappa Ba (IκBa) protein and increased the cellular IκBa level by reducing phosphorylation and promoting ubiquitination of the IκBa protein. Meanwhile, KCNQ1OT1 promoted the expression of IκBa by sponging miR-221. The effects of KCNQ1OT1 knockdown on promoting VSMC proliferation, migration, and secretion of inflammatory factors were abolished by IκBa overexpression. The roles of KCNQ1OT1 in reducing the intimal area and inhibiting IκBa expression were proved in the VG mouse model after KCNQ1OT1 overexpression. In conclusion, KCNQ1OT1 attenuated intimal hyperplasia by suppressing the inflammation and proliferation of VSMCs, in which the mechanism upregulated IκBa expression by binding to the IκBa protein and sponging miR-221.

6.
J Interferon Cytokine Res ; 35(3): 157-67, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25347351

RESUMO

Melanoma differentiation-associated gene-7 (mda-7)/interleukin-24 (IL-24) induces caspase-3 cleavage and subsequent activation via the intrinsic or extrinsic pathway to result in cancer cell-selective apoptosis, but whether mda-7/IL-24 may directly regulate caspase-3 through the post-translational modification remains unknown. Here, we reported that tumor-selective replicating adenovirus ZD55-IL-24 led to caspase-3 denitrosylation and subsequent activation, indicating that caspase-3 denitrosylation played a crucial role in ZD55-IL-24-induced cancer cell apoptosis. To confirm the relationship between caspase-3 denitrosylation and its activation in response to ZD55-IL-24, we treated carcinoma cells with the different nitric oxide (NO) regulators to modulate caspase-3 denitrosylation level, then observed the corresponding caspase-3 cleavage. We found that NO inhibitor 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxy-3-oxide (PTIO) promoted caspase-3 denitrosylation and caspase-3 cleavage, thereby exacerbating ZD55-IL-24-induced cancer cell apoptosis, whereas NO donor sodium nitroprusside (SNP) showed the opposite effect. Moreover, caspase-3 denitrosylation facilitated its downstream target poly ADP-ribose polymerase (PARP) degradation that further increased the apoptotic susceptibility. Although caspase-3 activation controlled by denitrosylation modification has emerged as an important regulator of programmed cell death, the detailed molecular mechanism by which caspase-3 exerts its denitrosylation modification in response to ZD55-IL-24 still needs to be elucidated. Thus, our results demonstrated that ZD55-IL-24 increased Fas expression to enhance thioredoxin reductase 2 (TrxR2), which was responsible for caspase-3 denitrosylation. Collectively, these findings elucidate that ZD55-IL-24 induces caspase-3 denitrosylation through Fas-mediated TrxR2 enhancement, thereby facilitating caspase-3 cleavage and the downstream caspase signaling pathway activation, which provides a novel insight into ZD55-IL-24-induced cancer-specific apoptosis by post-translational modification of the apoptotic executor caspase-3.


Assuntos
Caspase 3/metabolismo , Interleucinas/metabolismo , Adenoviridae/genética , Apoptose , Clonagem Molecular , Técnicas de Transferência de Genes , Células HeLa , Humanos , Interleucinas/genética , Especificidade de Órgãos , Processamento de Proteína Pós-Traducional/genética , Transdução de Sinais/genética , Tiorredoxina Redutase 2/metabolismo , Transgenes/genética , Receptor fas/metabolismo
7.
J Cancer Res Clin Oncol ; 139(11): 1845-52, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24005418

RESUMO

PURPOSE: To investigate the prognostic value of tumor-infiltrating regulatory T cells (Tregs) in the distribution of cancer nest, cancer stroma and normal mucosa and FOXP3-positive cancer cells in colon cancer patients after resection. METHODS: Paraffin blocks of operation resection of primary adenocarcinoma of colon were obtained from ninety patients. The distribution of tumor-infiltrating Tregs was detected by tissue microarray and immunohistochemistry staining technique to evaluate the prognostic effects by Kaplan-Meier and Cox regression analysis using median values as cutoff. RESULTS: The intratumoral Tregs counts were significantly higher than that in corresponding normal mucosa tissues (P < 0.001); the Tregs counts in cancer nest were significantly lower than that in corresponding cancer stroma tissues (P < 0.001); the increased intratumoral Tregs counts were associated with favorable prognosis (P < 0.05); the presence of Tregs in cancer nest was associated with unfavorable prognosis and was an independent prognostic factor for overall survival (P < 0.05). The appearance of FOXP3-positive cancer cells was associated with worse prognosis (P < 0.05). In addition, the frequency of the presence of FOXP3-positive cancer cells was higher in patients with lymphatic invasion (P < 0.001) and lower in patients with early TNM stage (P < 0.01). CONCLUSIONS: The higher tumor-infiltrating Tregs counts are closely associated with the improved prognostic effects of colon carcinoma. Tregs play different roles in cancer nest and cancer stroma. And the appearance of Tregs in cancer nest is a promising independent risk factor for overall survival in colon carcinoma. FOXP3-positive cancer cells may also be a risk factor for overall survival in colon carcinoma.


Assuntos
Neoplasias do Colo/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T Reguladores/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Idoso , Neoplasias do Colo/patologia , Feminino , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/imunologia , Humanos , Imuno-Histoquímica , Metástase Linfática , Linfócitos do Interstício Tumoral/patologia , Masculino , Gradação de Tumores , Estadiamento de Neoplasias , Inclusão em Parafina , Prognóstico , Fatores de Risco , Linfócitos T Reguladores/patologia , Análise Serial de Tecidos
8.
J Immunoassay Immunochem ; 33(4): 352-68, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22963485

RESUMO

RGD peptide (Arg-Gly-Asp tripeptide) binds to integrin αVß(3) and αVß(5), which is selectively expressed in tumor neovasculature and on the surface of some tumor cells. Some studies showed that coupling the RGD peptides to anticancer drugs yielded compounds with increased efficiency against tumors and lowered toxicity to normal tissues. The melanoma differentiation-associated gene-7/interleukin-24 gene (mda-7/IL-24) is a novel tumor-suppressor/cytokine gene that exhibits potent tumor-suppressive activity without damaging normal cells. To enhance the antitumor effect, we inserted a glycine residue into the wild type (mda-7/IL-24) between (164)Arg and (165)Asp to form a RGD peptide, named RGD-mda-7, then expressed RGD-mda-7 in Escherichia coli. Herein, we describe the expression and purification of RGD-mda-7. We detected the characterizations of immunostimulatory activity, tumor targeting, potent cytopathic effect, and apoptosis inducing exploited by RGD-mda-7 in tumor cells, and also compared these characterizations with wtmda-7/IL-24. The data showed that RGD-mda-7 had more potent tumor targeting and apoptosis-inducing effects than wtmda-7/IL-24.


Assuntos
Antineoplásicos/farmacologia , Interleucinas/imunologia , Oligopeptídeos/farmacologia , Antineoplásicos/isolamento & purificação , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clonagem Molecular , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imunização , Integrina alfaVbeta3/imunologia , Interleucinas/genética , Interleucinas/isolamento & purificação , Células MCF-7 , Mutação , Oligopeptídeos/genética , Oligopeptídeos/isolamento & purificação , Receptores de Vitronectina/imunologia , Relação Estrutura-Atividade
9.
J Interferon Cytokine Res ; 32(2): 66-73, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22248086

RESUMO

Melanoma differentiation-associated gene-7 (mda-7)/interleukin-24 (IL-24) has shown potent tumor cell apoptosis inducing capacity in multiple cancers. However, the apoptosis induction capacity of mda-7/IL-24 was low and directly correlated with the adhesion to tumor cells.Cell adhesion molecule integrin α(v)ß(3) expressed on the surface of several types of solid tumor cells, and they bind to arginine-glycine-aspartic acid (RGD) which enhanced the adhesion to tumor cells. This rout was exploited to construct a tumor-targeting gene RGD-IL-24 which can express RGD-MDA-7/IL-24 protein that includes the cell adhesive sequence (164)Arg-(165)Gly-(166)Asp (A Glycine residue was inserted into the recombinant MDA-7/IL-24 between Arg164 and Asp165 to form a RGD motif). We successfully got the MDA-7/IL-24 mutant by overlapping polymerase chain reaction (PCR) and evaluated its therapeutic efficacy for tumor cell lines MCF-7, HeLa, HepG2, and normal human lung fibroblast (NHLF) line. And we found that the expression of pCDNA3.1/RGD-IL-24 was same to the expression of pCDNA3.1/IL-24. The RGD-IL-24 enhanced the apoptosis-inducing function in tumor cells, but not in normal cells. In tumor cell lines, the apoptosis-inducing activities of RGD-IL-24 was significantly higher than IL-24 detecting by MTT assay, Annexin V, and Hoechst 33258 analysis. Further, pCDNA3.1/RGD-IL-24 showed a significant increase in the ratio of pro-apoptotic (bax) to anti-apoptotic (bcl-2) proteins in tumor cell lines, but not in NHLF cell line. Together, these results suggest that RGD-IL-24 can enhance the apoptosis of tumor cells and may provide a promising drug in tumor therapy.


Assuntos
Apoptose , Regulação Neoplásica da Expressão Gênica , Interleucinas/biossíntese , Mutação , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Oligopeptídeos , Adesão Celular/genética , Linhagem Celular Tumoral , Células HeLa , Células Hep G2 , Humanos , Integrina alfa5/biossíntese , Integrina alfa5/genética , Integrina beta3/biossíntese , Integrina beta3/genética , Interleucinas/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
10.
Cancer Biother Radiopharm ; 26(5): 647-55, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21902545

RESUMO

The melanoma differentiation-associated gene-7/interleukin-24 gene (mda-7/IL-24) is a novel tumor-suppressor/cytokine gene that exhibits potent tumor-suppressive activity without damaging normal cells. To enhance the antitumor effect, an mda-7/IL-24 mutant, RGD-mda-7, which includes the cell adhesive sequence 164Arg-165Gly-166Asp (RGD motif), was constructed and evaluated for bioactivity. RGD peptide binds to integrins α(V)ß(3) and α(V)ß(5), which are selectively expressed in tumor neovasculature and in the surface of some tumor cells. The wtmda-7/IL-24 and RGD-mda-7 were expressed in Escherichia coli and then purified and renatured. The immunostimulatory activity of RGD-mda-7 was assayed by stimulating peripheral blood mononuclear cells. The results suggested that the abilities of RGD-mda-7 to induce IL-6, TNF-α, and IFN-γ production were higher than wtmda-7/IL-24. Tumor targeting of RGD-mda-7 was assayed using cell adhesion experiments. The antitumor effect of the purified RGD-mda-7 on cell proliferation in vitro was evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) uptake, cell apoptosis by staining with fluorescent probes of FITC-annexin V and DAPI, and caspase-3 expression and activity. The in vitro results showed that RGD-mda-7 inhibited the proliferation of multiple tumor cell lines (Hela, ACHN, HepG2, and A549). Staining with fluorescent probes of FITC-annexin V and DAPI indicated that RGD-mda-7 could induce apoptosis more effectively in four tumor cell lines than wtmda-7/IL-24, but has no effect on normal cell line NHLF. Western blotting showed that treatment of tumor cells with RGD-mda-7 could activate apoptotic pathway by cleavage of caspase-3 as same as wtmda-7/IL-24. Further, RGD-mda-7 group showed a higher cleaved level of caspase-3, but not in NHLF cells. These results demonstrate that RGD-MDA-7 possesses more potent antitumor effects than wtmda-7/IL-24 and therefore merits further investigation in preclinical and clinical studies.


Assuntos
Integrinas/metabolismo , Interleucinas/farmacologia , Melanoma/tratamento farmacológico , Oligopeptídeos/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Apoptose/efeitos dos fármacos , Processos de Crescimento Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células HeLa , Humanos , Interferon gama/biossíntese , Interleucina-6/biossíntese , Interleucinas/genética , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Terapia de Alvo Molecular , Oligopeptídeos/genética , Proteínas Recombinantes de Fusão/genética , Fator de Necrose Tumoral alfa/biossíntese
11.
Tumour Biol ; 32(2): 273-83, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20963645

RESUMO

Ki-67 plays a crucial role in cell proliferation as well as maintenance or regulation of cell division. The mechanism governing the Ki-67 gene expression remains unknown. Thus, we cloned the core promoter of the human Ki-67 gene and further investigated its transcriptional regulation. The putative Sp1 binding sites were confirmed by electrophoretic mobility shift assay together with an anti-Sp1 antibody-mediated supershift assay. Deletion mutagenesis and firefly luciferase reporter gene assay demonstrated the essential contribution of Sp1 on transcriptional activation of the Ki-67 gene. In this study, we first confirm that there are three Sp1 binding sites in the Ki-67 core promoter. Two Sp1 sites (one at position -159 to -145 nt and the other at position -14 to +12 nt) are mainly involved in transcriptional regulation of the Ki-67 gene. Overexpression of Sp1 can enhance the Ki-67 promoter activity. However, down-regulation of Sp1 expression using siRNA-Sp1 and mithramycin effectively inhibits the Ki-67 gene transcription. Our results suggest that Sp1 is essential for basal promoter activity of the human Ki-67 gene. Inhibition of the Ki-67 transcriptional activity through abolishment of Sp1 may provide the useful prospect for gene therapy.


Assuntos
Adenocarcinoma/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Antígeno Ki-67/metabolismo , Neoplasias Pulmonares/metabolismo , Fator de Transcrição Sp1/fisiologia , Neoplasias do Colo do Útero/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/fisiopatologia , Sequência de Bases , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Antígeno Ki-67/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/fisiopatologia , Dados de Sequência Molecular , Inibidores da Síntese de Ácido Nucleico/farmacologia , Plicamicina/farmacologia , RNA Interferente Pequeno/farmacologia , Fator de Transcrição Sp1/genética , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/fisiopatologia
12.
Tumour Biol ; 32(1): 63-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20711822

RESUMO

Peptide-based immunotherapy strategies appear promising as an approach to successfully induce an antitumor immune response and prolong survival in patients with various cancers. Protein antigens and their specific epitopes are formulation targets for anti-tumor vaccines. Bioinformatical approaches to predict major histocompatibility complex binding peptides can facilitate the resource-consuming effort of T cell epitope identification. Proliferating cell nuclear antigen including Ki-67 and PCNA, associated with the proliferation process of the cell, seems to be an attractive new target for tumor-specific immunotherapy. In this study, we predicted seven HLA-A*0201-restricted CTL candidate epitope of Ki-67 and eight epitope of PCNA by computer algorithm SYFPEITHI, BIMAS, and IEDB_ANN. Subsequently, biological functions of these peptides were tested by experiments in vitro. We found Ki-67((280-288)) (LQGETQLLV) had the strongest binding-affinity with HLA-A*0201. Further study revealed that Ki-67((280-288)) increased the frequency of IFN-γ-producing T cells compared to a negative peptide. Because Ki-67 was broadly expressed in most advanced malignant tumors, indicating a potential anti-tumor application in the future.


Assuntos
Epitopos de Linfócito T/imunologia , Antígenos HLA-A/imunologia , Antígeno Nuclear de Célula em Proliferação/imunologia , Linfócitos T Citotóxicos/imunologia , Neoplasias da Mama/sangue , Neoplasias da Mama/imunologia , Neoplasias Colorretais/sangue , Neoplasias Colorretais/imunologia , Bases de Dados Factuais , Ensaio de Imunoadsorção Enzimática , Feminino , Antígeno HLA-A2 , Humanos , Antígeno Ki-67/imunologia , Fragmentos de Peptídeos/imunologia , Neoplasias Gástricas/sangue , Neoplasias Gástricas/imunologia
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 26(3): 261-3, 266, 2010 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-20230693

RESUMO

AIM: To explore the effect of nimesulide on human gammadeltaT cell function. METHODS: gammadeltaT cells were cultured routinely, collected on the 9th day, and then induced with nimesulide at indicated concentrations (0.25 micromol/L, 0.5 micromol/L, 1 micromol/L, 2 micromol/L, 4 micromol/L, respectively). Twenty-four hours after induction, the supernatants were collected to detect IFN-gamma, TNF-alpha and IL-12, whereas the cells were assayed for perforin, granzyme B and NKG2D by flow cytometry (FCM) and for killing of gastric cancer cells by LDH. RESULTS: Nimesulide (1 micromol/L) caused gammadeltaT cells to express more of perforin and granzyme B (62.8% and 72.7%, respectively) than the control group (51.4% and 60.9%, respectively) (P<0.05). Likewise, nimesulide (1 micromol/L) enabled gammadeltaT cells to secret more of IFN-gamma and TNF-alpha (262.3 ng/L and 177.5 ng/L, respectively) than the control group (196.1 ng/L and 158.5 ng/L, respectively) (P<0.05). Nimesulide did not affect IL-12 secreting capability of gammadeltaT cells as compared with the control group (P>0.05). Nimesulide-stimulated gammadeltaT cells killed more of SGC-7901 and BCG-823 gastric cancer cells (73% and 70%, respectively) than the control group(54% and 53%, respectively) (P<0.05). CONCLUSION: Nimesulide made gammadeltaT cells to express more perforin and granzyme B and to secret more IFN-gamma and TNF-alpha into the supernatant, leading to higher killing rate of SGC-7901 and BCG-823 gastric cancer cells. The above data provides experimental basis on the clinical use of nimesulide to prevent and treat digestive tract tumors.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Perforina/efeitos dos fármacos , Neoplasias Gástricas/imunologia , Sulfonamidas/farmacologia , Subpopulações de Linfócitos T/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Granzimas/imunologia , Humanos , Interferon gama/imunologia , Perforina/imunologia , Subpopulações de Linfócitos T/enzimologia , Fator de Necrose Tumoral alfa/imunologia
14.
Cancer Biol Ther ; 8(7): 599-606, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19305163

RESUMO

It has been demonstrated that interleukin 18 (IL-18) exerts antitumor activity. In this study, we investigated whether oncolytic adenovirus-mediated gene transfer of IL-18 could induce strong antitumor activity. A tumor-selective replicating adenovirus expressing IL-18 (ZD55-IL-18) was constructed by insertion of an IL-18 expression cassette into the ZD55 vector, which is based on deletion of the adenoviral E1B 55-kDa gene. ZD55-IL-18 could express substantially more IL-18 than Ad-IL-18 because of replication of the vector. It has been shown that ZD55-IL-18 exerted a strong cytopathic effect and significant apoptosis in renal cell carcinoma. ZD55-IL-18 significantly decreased VEGF and CD34 expression in the tumor cells. Treatment of established tumors with ZD55-IL-18 showed much stronger antitumor activity than that induced by ZD55-EGFP or Ad-IL-18. These data indicated that oncolytic adenovirus expressing IL-18 could exert potential antitumor activity via inhibition of angiogenesis and offer a novel approach to cancer therapy.


Assuntos
Carcinoma de Células Renais/terapia , Terapia Genética/métodos , Interleucina-18/genética , Neoplasias Renais/terapia , Adenoviridae/genética , Adenoviridae/fisiologia , Animais , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/virologia , Linhagem Celular Tumoral , Vetores Genéticos/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Interleucina-18/biossíntese , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/genética , Neoplasias Renais/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Terapia Viral Oncolítica , Células Tumorais Cultivadas , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cancer Biol Ther ; 8(1): 84-91, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19029834

RESUMO

RNA interference (RNAi) has been proved to be a powerful tool for gene knockdown purpose and holds a great promise for the treatment of cancer. Our previous study demonstrated that the reduction of hTERT expression by means of chemically synthesized siRNAs and shRNAs expressed from plasmid resulted in proliferation inhibition in human renal carcinoma cells. In this study, we constructed a novel oncolytic adenovirus-based shRNA expression system, ZD55-hTERT, and to explore ZD55-hTERT-mediated RNAi for hTERT gene silencing. Our results showed that ZD55-hTERT could induce silencing of hTERT gene effectively, allow for efficient tumor-specific viral replication and induce the apoptosis of tumor cells effectively in vitro and in nude mice. We conclude that combining shRNA gene therapy and oncolytic virotherapy can enhance antitumor efficacy as a result of synergism between CRAd oncolysis and shRNA antitumor responses.


Assuntos
Adenoviridae/genética , Neoplasias Renais/genética , Neoplasias Renais/patologia , Interferência de RNA/fisiologia , Telomerase/genética , Divisão Celular , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Humanos , Neoplasias Renais/enzimologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Plasmídeos/genética , RNA Interferente Pequeno/genética , Vacinas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...