Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(1): 74-79, 2024 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-38246180

RESUMO

Tumor aerobic glycolysis is one of the main features of tumor metabolic reprogramming. This abnormal glycolytic metabolism provides bioenergy and biomaterials for tumor growth and proliferation. It is worth noting that aerobic glycolysis will not only provide biological materials and energy for tumor cells, but also help tumor cells to escape immune surveillance through regulation of immune microenvironment, thereby resisting tumor immunotherapy and promoting tumor progression. Based on the pathogenesis of renal cell carcinoma, this paper describes the characteristics of aerobic glycolysis, the effect of glycolytic metabolism on the immune microenvironment of renal cell carcinoma, the effect of glycolysis inhibitors on the immune microenvironment of renal cell carcinoma, and the prospect of glycolysis inhibitors combined with immune checkpoint inhibitors in the treatment of renal cell carcinoma.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Carcinoma de Células Renais/terapia , Imunoterapia , Glicólise , Reprogramação Metabólica , Neoplasias Renais/terapia , Microambiente Tumoral
2.
Int J Biol Macromol ; 258(Pt 1): 128848, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38114003

RESUMO

The survival benefit for patients with gastric cancer (GC) is modest due to its high transfer potential. Targeted therapy for metastasis-related genes in GC may be a viable approach, however, inhibitors specifically targeting GC are limited. In this study, GC patient-derived xenografts (PDX) with metastatic burden were established via orthotopic transplantation. PCR-Array analysis of primary and metastatic tumors revealed EPH receptor B2 (EPHB2) as the most significantly upregulated gene. The interaction between the EPHB2 receptor and its cognate-specific EFNB1 ligands was high in GC and correlated with a poor prognosis. Fc-EFNB1 treatment increased the invasion and migration abilities of GC cells and induced a high EPHB2 expression. EPHB2 knockdown in GC cells completely abolished the ephrin ligand-induced effects on invasion and migration abilities. Signal transduction analysis revealed Wnt/ß-catenin and FAK as downstream signaling mediators potentially inducing the EPHB2 phenotype. In conclusion, the observed deregulation of EPHB2/EFNB1 expression in GC enhances the invasive phenotype, suggesting a potential role of EPHB2/EFNB1 compound in local tumor cell invasion and the formation of metastasis.


Assuntos
Receptor EphB2 , Neoplasias Gástricas , Humanos , Receptor EphB2/genética , Receptor EphB2/metabolismo , Neoplasias Gástricas/patologia , Efrina-B1/genética , Efrina-B1/metabolismo , beta Catenina/metabolismo , Ligantes , Via de Sinalização Wnt , Movimento Celular/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética
3.
Front Immunol ; 14: 1219652, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37457713

RESUMO

Background: Alterations in Mitochondrial DNA methylation (MTDM) exist in many tumors, but their role in breast cancer (BC) development remains unclear. Methods: We analyzed BC patient data by combining scRNA-seq and bulk sequencing. Weighted co-expression network analysis (WGCNA) of TCGA data identified mitochondrial DNA methylation (MTDM)-associated genes in BC. COX regression and LASSO regression were used to build prognostic models. The biological function of MTDM was assessed using various methods, such as signaling pathway enrichment analysis, copynumber karyotyping analysis, and quantitative analysis of the cell proliferation rate. We also evaluated MTDM-mediated alterations in the immune microenvironment using immune microenvironment, microsatellite instability, mutation, unsupervised clustering, malignant cell subtype differentiation, immune cell subtype differentiation, and cell-communication signature analyses. Finally, we performed cellular experiments to validate the role of the MTDM-associated prognostic gene NCAPD3 in BC. Results: In this study, MTDM-associated prognostic models divided BC patients into high/low MTDM groups in TCGA/GEO datasets. The difference in survival time between the two groups was statistically significant (P<0.001). We found that high MTDM status was positively correlated with tumor cell proliferation. We analyzed the immune microenvironment and found that low-MTDM group had higher immune checkpoint gene expression/immune cell infiltration, which could lead to potential benefits from immunotherapy. In contrast, the high MTDM group had higher proliferation rates and levels of CD8+T cell exhaustion, which may be related to the secretion of GDF15 by malignant breast epithelial cells with a high MTDM status. Cellular experiments validated the role of the MTDM-associated prognostic gene NCAPD3 (the gene most positively correlated with epithelial malignant cell proliferation in the model) in BC. Knockdown of NCAPD3 significantly reduced the activity and proliferation of MDA-MB-231 and BCAP-37 cells, and significantly reduced their migration ability of BCAP-37 cell line. Conclusion: This study presented a holistic evaluation of the multifaceted roles of MTDM in BC. The analysis of MTDM levels not only enables the prediction of response to immunotherapy but also serves as an accurate prognostic indicator for patients with BC. These insightful discoveries provide novel perspectives on tumor immunity and have the potentially to revolutionize the diagnosis and treatment of BC.


Assuntos
Neoplasias da Mama , DNA Mitocondrial , Humanos , Feminino , DNA Mitocondrial/genética , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Análise da Expressão Gênica de Célula Única , Metilação de DNA , Prognóstico , Imunoterapia , Microambiente Tumoral/genética
4.
Cell Commun Signal ; 21(1): 159, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37370081

RESUMO

Cancer stem cells (CSCs) are considered to be responsible for tumor recurrence and metastasis. Therefore, clarification of the mechanisms involved in CSC stemness maintenance and cell fate determination would provide a new strategy for cancer therapy. Unregulated cellular energetics has been accepted as one of the hallmarks of cancer cells, but recent studies have revealed that mitochondrial metabolism can also actively determine CSC fate by affecting nuclear stemness gene expression. Herein, from the perspective of mito-nuclear communication, we review recent progress on the influence of mitochondria on CSC potential from four aspects: metabolism, dynamics, mitochondrial homeostasis, and reactive oxygen species (ROS). Video Abstract.


Assuntos
Mitomicina , Recidiva Local de Neoplasia , Humanos , Recidiva Local de Neoplasia/patologia , Diferenciação Celular , Mitomicina/metabolismo , Mitocôndrias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
5.
Cancer Lett ; 563: 216188, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37076041

RESUMO

Monoamine oxidase A (MAOA) is a mitochondrial enzyme that catalyzes the oxidative deamination of monoamine neurotransmitters and dietary amines. Previous studies have shown that MAOA is clinically associated with prostate cancer (PCa) progression and plays a key role in almost each stage of PCa, including castrate-resistant prostate cancer, neuroendocrine prostate cancer, metastasis, drug resistance, stemness, and perineural invasion. Moreover, MAOA expression is upregulated not only in cancer cells but also in stromal cells, intratumoral T cells, and tumor-associated macrophages; thus, targeting MAOA can be a multi-pronged approach to disrupt tumor promoting interactions between PCa cells and tumor microenvironment. Furthermore, targeting MAOA can disrupt the crosstalk between MAOA and the androgen receptor (AR) to restore enzalutamide sensitivity, blocks glucocorticoid receptor (GR)- and AR-dependent PCa cell growth, and is a potential strategy for immune checkpoint inhibition, thereby alleviating immune suppression and enhancing T cell immunity-based cancer immunotherapy. MAOA is a promising target for PCa therapy, which deserves further exploration in preclinical and clinical settings.


Assuntos
Monoaminoxidase , Neoplasias da Próstata , Masculino , Humanos , Monoaminoxidase/metabolismo , Monoaminoxidase/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Próstata/patologia , Proliferação de Células , Receptores Androgênicos , Linhagem Celular Tumoral , Microambiente Tumoral
6.
Oncol Lett ; 23(4): 138, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35317027

RESUMO

Salinomycin (SAL), a typical ion carrier antibiotic, inhibits tumor growth and metastasis by inducing apoptosis or autophagy in cancer or cancer stem cells and thus overcomes drug resistance. 17-allylamino-17-demethoxygeldanamycin (17-AAG), a heat shock protein Hsp90 competitive inhibitor, also has a role in inhibiting tumor development. However, their combination on the growth of breast cancer cells and its specific mechanism remains to be elucidated. The present study tested the influence of SAL and 17-AAG on cell growth, apoptosis and autophagy by MTT assays, Annexin V-FITC and propidium iodide double staining assay and immunoelectron microscopy. The influence of SAL and 17-AAG on proteomics was investigated by isobaric tag for relative and absolute quantitation. It was found that SAL combined with 17-AAG synergistically inhibited the cell growth and induced the apoptosis in a concentration-dependent manner, with the expression of caspase 3 and Bcl-2 were decreased while the expression of Bax was increased. In addition, SAL combined with 17-AAG inhibited autophagy, with the expression of microtubule-associated protein 1 light chain 3, Beclin1, p62 being decreased. Mechanistically, SAL combined with 17-AAG synergistically inhibited the reactive oxygen species/JNK signaling pathway. In conclusion, SAL combined with 17-AAG had a synergistic inhibitory effect on cell growth of breast cancer via inducing apoptosis and inhibiting autophagy. The present study might provide a new strategy for potential clinical application of SAL as a new anti-tumor drug especially as a drug combination with other molecular targeting therapeutics.

7.
Biochem Biophys Res Commun ; 606: 135-141, 2022 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-35349822

RESUMO

Neuroendocrine transdifferentiation (NED) of prostate cancer (PCa) is the main cause of failure of androgen receptor inhibitor treatment. However, the molecular mechanisms underlying the development of NEPC, especially treatment-induced NEPC, remain unclear. Emerging evidence indicates that elevated monoamine oxidase A (MAOA) contribute to the proliferation, cell stemness, and bone metastasis in PCa. Here, we generated an enzalutamide-induced NED cell model to assess the role of MAOA during NED. Overall, MAOA expression was significantly increased upon Enz long-term exposure and was required for neuroendocrine marker expression. In particular, Enz was found to induce NED via the MAOA/mTOR/HIF-1α signaling axis. Further analyses revealed that the MAOA inhibitor clorgyline(CLG) may bring multiple benefits to CRPC patients, including better therapeutic effect and delays NED. These findings suggest that MAOA may be an important target for the development of anti-NED therapies, thereby providing a novel strategy for the combined application of CLG and AR inhibitors in the clinic.


Assuntos
Transdiferenciação Celular , Monoaminoxidase , Neoplasias da Próstata , Linhagem Celular Tumoral , Humanos , Masculino , Monoaminoxidase/genética , Monoaminoxidase/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais
8.
Lab Invest ; 102(4): 332-340, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34937865

RESUMO

Prostate cancer is the most common cancer among men and has a high incidence and associated mortality worldwide. It is an androgen-driven disease in which tumor growth is triggered via ligand-mediated signaling through the androgen receptor (AR). Recent evidence suggests that the widespread use of effective AR pathway inhibitors may increase the occurrence of neuroendocrine prostate cancer (NEPC), an aggressive and treatment-resistant AR-negative variant; however, mechanisms controlling NEPC development remain to be elucidated. Various preclinical models have recently been developed to investigate the mechanisms driving the NEPC differentiation. In the present study, we summarized strategies for the development of NEPC models and proposed a novel method for model evaluation, which will help in the timely and accurate identification of NEPC by virtue of its ability to recapitulate the heterogeneity of prostate cancer. Moreover, we discuss the origin and the mechanism of NEPC. The understanding of the regulatory network mediating neuroendocrine differentiation presented in this review could provide valuable insights into the identification of novel drug targets for NEPC as well as into the causes of antiandrogenic drug resistance.


Assuntos
Carcinoma Neuroendócrino , Neoplasias da Próstata , Carcinoma Neuroendócrino/metabolismo , Carcinoma Neuroendócrino/patologia , Linhagem Celular Tumoral , Humanos , Masculino , Próstata/patologia , Neoplasias da Próstata/metabolismo , Transdução de Sinais
9.
Front Oncol ; 11: 673199, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996603

RESUMO

Immunotherapy has been used as a first-line treatment for a variety of advanced tumors, allowing remarkable progress to be made in cancer treatment. Nonetheless, only a small number of patients can benefit from immune checkpoint inhibitor monotherapy. To improve the effect of immunotherapy, the underlying mechanism of combination therapy was investigated in the context of an intact human tumor immune microenvironment using mice with a human immune system (HIS) bearing human tumors. Herein, we summarize and discuss strategies for the development and use of HIS mice models in tumor immunotherapies. Most importantly, this review proposes a method of t11umor identification and classification in HIS mice based on the tumor-infiltrating lymphocytes and PD-L1 expression, and according to this classification, we propose different combination treatment strategies that can be utilized to enhance the effect of immunotherapy. Thus, we provide effective experimental schemes for tumor immunotherapy in HIS mice models.

10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 35(6): 526-532, 2019 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-31292057

RESUMO

Objective To investigate the effect and mechanism of Trillium saponins on the invasion and migration of HuH-7 cells regulated by human ß-defensin 2 (HBD-2). Methods HuH-7 cells were treated with 0.5, 1.0, 2.0, 4.0 mg/L Trillium saponins. Cell proliferation was detected by MTT assay. TranswellTM chamber was used to measure cell invasion and migration. The levels of matrix metalloproteinase-2 (MMP2), HBD-2 and matrix metalloproteinase-9 (MMP9) were detected by Western blot analysis. HBD-2 small interfering RNA (siRNA) was transfected into HuH-7 cells. The interference effect of Trillium saponins treatment was verified by real-time quantitative PCR and Western blot analysis, and TranswellTM assay was used to detect cell invasion and migration. Results Except that 0.5 mg/L Trillium saponins had no effect on the proliferation of HuH-7 cells, the proliferation of HuH-7 cells was inhibited by all other doses of Trillium saponins. (0.5, 1.0) mg/L of Trillium saponins down-regulated the levels of MMP2 and MMP9 proteins, increased the level of HBD-2 protein and inhibited cell invasion, migration. HBD-2 siRNA transfection significantly reduced the level of HBD-2 in HuH-7 cells. Down-regulation of HBD-2 increased the invasive, migratory ability and increased the levels of MMP2 and MMP9 proteins in HuH-7 cells treated with Trillium saponins. Conclusion Trillium saponins can inhibit the invasion and migration of HuH-7 cells by promoting the expression of HBD-2.


Assuntos
Invasividade Neoplásica , Saponinas/farmacologia , Trillium/química , beta-Defensinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Compostos Fitoquímicos/farmacologia
11.
Oncol Lett ; 16(2): 1397-1404, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30008816

RESUMO

The present study aimed to investigate the effect of alkaloids and carbinol extracts from lily on the proliferation of SGC-7901 cells, as well as the underlying mechanism. SGC-7901 cells were incubated with different concentrations of alkaloid or carbinol extracts for 24, 48 or 72 h. MTT assays were used to measure the inhibition rate of SGC-7901 cell proliferation. Inverted phase contrast and fluorescence microscopy was used to observe morphological changes of SGC-7901 cells. Flow cytometry was employed to detect cell cycle progression and apoptosis rates of SGC-7901 cells. Western blotting was performed to measure the expression of caspase-3, Fas and Fas ligand (FasL) proteins in SGC-7901 cells. The inhibition rate of SGC-7901 cell proliferation was significantly enhanced with increasing drug concentrations and time elapsed. Treatment with alkaloid or carbinol extracts deteriorated the morphology of SGC-7901 cells in a dose-dependent manner. Alkaloid and carbinol extracts arrested SGC-7901 cells in the G2/M phase, and induced apoptosis in a dose-dependent manner. Alkaloid and carbinol extracts enhanced caspase-3, and Fas expression, but reduced FasL expression in SGC-7901 cells. The present study demonstrated that alkaloids and carbinol extracts from lily inhibited the proliferation of gastric carcinoma SGC-7901 cells by arresting cells in the G2/M phase. The upregulation of caspase-3 and Fas proteins, and the downregulation of FasL protein may be an important mechanism for the induction of SGC-7901 cell apoptosis.

12.
Mol Med Rep ; 16(2): 1063-1070, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627587

RESUMO

The aim of the present study was to investigate the effects and mechanisms of 17­AAG combined with salinomycin treatment on proliferation and apoptosis of the SGC­7901 gastric cancer cell line. An MTT assay was used to detect the proliferation of SGC­7901 cells. Morphological alterations of cells were observed under inverted phase­contrast and fluorescence microscopes. Cell cycle and apoptosis were assessed by flow cytometry analysis. The protein expression of nuclear factor (NF)­κB p65 and Fas­ligand (L) were evaluated by immunocytochemistry. Salinomycin with a concentration range of 1­32 µmol/l was demonstrated to inhibit growth of SGC­7901 cells effectively, affect the morphology and apoptosis rate of cells, and arrest SGC­7901 cells in S phase. Furthermore, salinomycin significantly increased the protein expression of Fas­L and decreased the protein expression of NF­κB p65. The alterations in SGC­7901 cells co­treated with salinomycin and 17­AAG were more significant compared with cells treated with one drug only. In conclusion, the individual use of salinomycin and combined use with 17­AAG may significantly inhibit SGC­7901 gastric cancer cell proliferation and induce cell apoptosis. The potential mechanisms may be associated with upregulation of Fas­L and downregulation of NF­κB. These results provide a basis for the potential use of salinomycin in gastric cancer treatment.


Assuntos
Benzoquinonas/farmacologia , Lactamas Macrocíclicas/farmacologia , Piranos/farmacologia , Neoplasias Gástricas/patologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Proteína Ligante Fas/metabolismo , Humanos , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Receptor fas/metabolismo
13.
Mol Med Rep ; 16(2): 1031-1038, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627601

RESUMO

The present study aimed to investigate the anticancer effects of cisplatin (DDP) combined with salinomycin (SAL) on the gastric cancer cell line SGC­7901, as well as to explore the mechanisms underlying their actions. An MTT assay was used to evaluate the inhibitory effects of SAL, DDP and their combination on gastric cancer cell proliferation. Morphological alterations of cancer cells following treatment were observed under an inverted phase­contrast microscope and a fluorescence microscope. Cell cycle progression and apoptosis were analyzed using flow cytometry. The expression of nuclear factor (NF)­κB p65 and Fas protein ligand (L) in cancer cells was assessed using immunocytochemistry. The present results demonstrated that the combination of SAL and DDP significantly inhibited the proliferation (P<0.05) and altered the morphological characteristics of SGC­7901 cells, thus suggesting that SAL may enhance the susceptibility of gastric cancer cells to DDP. In addition, treatment with a combination of SAL and DDP resulted in S phase­arrest and increased the apoptotic rate of SGC­7901 cells. Furthermore, marked FasL upregulation and NF­κB p65 downregulation were observed in cancer cells treated with the combination of SAL and DDP. The results of the present study demonstrated that the combination of SAL and DDP induced the apoptosis of human gastric cancer cells, and suggested that the underlying mechanism may involve the upregulation of FasL and downregulation of NF­κB p65.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cisplatino/uso terapêutico , Piranos/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Cisplatino/farmacologia , Proteína Ligante Fas/metabolismo , Humanos , Piranos/farmacologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fator de Transcrição RelA/metabolismo
14.
Int J Clin Exp Med ; 8(9): 15840-5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26629085

RESUMO

This study is to investigate the therapeutic effects of bone marrow mesenchymal stem cells (BMSCs) expressing interleukin (IL)-12 on malignant ascites tumor-bearing mice and the related mechanisms. Malignant ascites tumor mouse model was established by the intraperitoneal inoculation with MethA or H22 tumor cells. Mouse BMSCs were transfected with lentiviral vector containing IL-12, and then transplanted into these mouse models via intraperitoneal injection. The peritoneal permeability in these mice was evaluated and compared. The contents of INF-γ and VEGF in ascites were determined by ELISA. Mouse models receiving IL-12-expressing BMSCs were rechallenged with tumor cells, and the animal survival was observed and analyzed. In both MethA and H22 tumor cell-induced malignant ascites tumor mouse models, there were no significant differences in the peritoneal permeability between the normal saline (NS), BMSC-control, and BMSC-null groups. However, compared with NS control group, the peritoneal permeability was significantly decreased by IL-12-expressing BMSCs. Moreover, ELISA showed that, in both the MethA and H22 tumor cell-induced mouse models, compared with the NS control group, the contents of INF-γ in ascites were significantly elevated, while the contents of VEGF in ascites were significantly decreased, in the BMSC-IL-12 groups. In addition, IL-12-expressing BMSCs significantly elongated the survival of mouse models after rechallenging with tumor cells. IL-12-expressing BMSCs exert protective effects against malignant ascites tumor, and the anti-tumor effects might be associated with the enhanced anti-tumor immunity. Our findings might bring new insights into the treatment of tumors with immunotherapy.

15.
Exp Ther Med ; 8(4): 1330-1334, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25187849

RESUMO

The objective of the present study was to investigate the effects of mesenchymal stem cells (MSCs) genetically modified by lentivirus-mediated mouse interleukin-12 (Lenti-mIL-12) in treating malignant ascites in mice. The in vitro chemotactic effect of Lenti-mIL-12-MSC culture supernatant on dendritic cells was investigated using a chemotaxis chamber. Liver cancer H22 and MethA ascites models were constructed. Mice were divided evenly into four groups: Normal saline, MSC, Null and Lenti-mIL-12-MSC. The survival rate, ascites volume and red blood cell number were measured for these groups. The toxicity and side effects of Lenti-mIL-12-MSCs were investigated using visual and microscopy inspections. The results indicated that mIL-12 had a strong chemotactic effect on dendritic cells. mIL-12 was highly expressed in ascites of Lenti-mIL-12-MSC-treated mice. Lenti-mIL-12-MSCs reduced the volume of ascites and the number of red blood cells in ascites and thus increased the survival rate and prolonged the survival duration of the mice. Furthermore, Lenti-mIL-12-MSCs showed no toxicity and side effects on the mice with malignant ascites. In conclusion, the results demonstrated that Lenti-mIL-12-MSCs inhibited the growth of ascites and promoted the survival of tumor-bearing mice, suggesting that Lenti-mIL-12-MSCs exerts a therapeutic effect on malignant ascites by stimulating the immune responses of the mice.

16.
Zhonghua Xin Xue Guan Bing Za Zhi ; 42(3): 202-7, 2014 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-24831476

RESUMO

OBJECTIVE: To explore the association between LMNA gene mutation and familiar dilated cardiomyopathy (DCM) (FDCM) and idiopathic DCM (IDCM) in Uygurs and Hans people in Xinjiang area. METHODS: Peripheral blood samples were collected from 28 family member with FDCM and 123 sporadic patients with IDCM(56 Uygur patients and 67 Han patients), 80 Uygur and 80 Han people were chosen as normal controls. PCR was used to amplify the 12 exons of LMNA gene. The amplified products were sequenced and compared with the standard sequence in the NCBI to determine the mutation sites. RESULTS: Transmission of the allele C and T of rs4641 was similar in Han FDCM patients. One new variation(c.1714C>T) located at exon 10 of LMNA gene was identified in 1 Han patient with IDCM, this mutation caused an amino acid substitution (R572C). In Uygurs people, rs553016 polymorphism was significant different between IDCM and control groups (P < 0.05). Logistic regression revealed that rs553016 was an independent risk factor for Uygurs patients with IDCM (OR = 3.178, P = 0.035). CONCLUSIONS: LMNA rs4641 is not associated with FDCM of Hans people in Xinjiang while LMNA mutation is associated with IDCM and rs533106 polymorphism is an independent risk factor for Uygurs patients with IDCM.


Assuntos
Cardiomiopatia Dilatada/genética , Lamina Tipo A/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Cardiomiopatia Dilatada/etnologia , Estudos de Casos e Controles , Criança , Pré-Escolar , China , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Polimorfismo Genético , Fatores de Risco , Adulto Jovem
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(2): 271-5, 2013 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-23443787

RESUMO

OBJECTIVE: To study the effect of the HSP90 inhibitor, 17-allylamino-17-demethoxygeldanamycin (17-AAG), on cell proliferation and apoptosis of human cancer SGC-7901 cells and explore the mechanisms. METHODS: The inhibitory effect of 17-AAG on the proliferation and morphology of SGC-7901 cells was assessed with MTT assay and DNA-PI staining, respectively. Flow cytometry was employed to analyze the changes in cell cycle and apoptosis of the cells following 17-AAG exposure. The cellular expression of Fas protein was detected by immunohistochemistry. RESULTS: 17-AAG significantly suppressed the proliferation of SGC-7901 cells in a time- and dose-dependent manner. After treatment with 17-AAG for 48 h, SGC-7901 cells showed cell cycle arrested at G(2)/M stage, and the cell apoptosis rate increased with the 17-AAG concentration. The expression of Fas protein in the cytoplasm of SGC-7901 cells increased gradually with the increase of 17-AAG concentration. CONCLUSION: 17-AAG can induce apoptosis, alters the cell cycle distribution and up-regulates the expression of Fas protein in SGC-7901 cells to suppress the cell proliferation.


Assuntos
Apoptose/efeitos dos fármacos , Benzoquinonas/farmacologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Lactamas Macrocíclicas/farmacologia , Neoplasias Gástricas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Receptor fas/metabolismo
18.
Oncol Rep ; 27(2): 383-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22075844

RESUMO

Povidone-iodine (PVP-I) is widely used in clinical practice as an antiseptic and flushing agent after surgery to remove a tumor. Our present study was designed to determine whether diluted PVP-I is cytotoxic to colon cancer cells and ascetic tumor cells in vitro and to examine its antitumor effects in vivo. In vitro, CT26 and H22 cells treated with different concentrations of diluted PVP-I (0-1.56 µg/ml) were analyzed using the mononuclear cell direct cytotoxicity assay (MTT) and a flow cytometry assay. In vivo, Balb/c mice injected in the abdominal cavity with CT26 cells or H22 cells were treated intraperitoneally with different concentrations of PVP-I (0-312.5 µg/mouse), cisplatin (40 mg/kg) or 5'-FU (30 mg/kg) or left untreated. In vitro, the studies demonstrated the antiproliferative and significant apoptosis-inducing effects of PVP-I in a dose- and time-dependent manner. In vivo, PVP-I significantly repressed the growth of H22 and CT26 cells in Balb/c mice compared to controls. To explore the mechanism of the antitumor effect of PVP-I, the superoxide dismutase (SOD) activity of ascites extracted from a mouse model and the supernatant of CT26 cells was detected by an SOD kit. The activity of SOD was significantly inhibited in the experimental groups. Taken together, our data suggest that PVP-I exhibits a strong inhibitory effect on tumor growth in colon cancer (CT26) and hepatoma (H22) resulting from apoptosis, both in vitro and in vivo, suggesting a new potential therapeutic approach after tumor excision surgery to colon cancer and hepatoma.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias/enzimologia , Povidona-Iodo/farmacologia , Superóxido Dismutase/antagonistas & inibidores , Animais , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Povidona-Iodo/uso terapêutico , Povidona-Iodo/toxicidade
19.
Vaccine ; 29(25): 4218-24, 2011 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-21497630

RESUMO

Anti-angiogenesis represents an indispensible strategy for cancer therapy. As a strictly endothelial-specific adhesion molecule, vascular endothelial cadherin (VE-cadherin, VE-cad) is a promising anti-angiogenesis target. In this study a recombinant adenovirus vector modified with mannan was used to deliver VE-cad (AdVEC-m) and we tried to explore its feasibility as an antitumour agent in mouse cancer models. The immunogenic delivery of VE-cad resulted in obvious prophylactic and therapeutic inhibition of tumour growth and prolonged survival in mice. In the meantime angiogenesis declined apparently within the tumours measured by immunohistochemistry staining and coated alginate bead assay in vivo. Anti-VE-cad antibodies were identified by western blot analysis and enzyme-linked immunosorbent assay (ELISA). VE-cad-specific T lymphocyte cytotoxicity responses (CTL) were detected by chromium (51Cr) release assay of splenocytes from AdVEC-m treated mice. These results demonstrate that mannan modification is able to enhance antigen delivery and immune responses, and the way of immunogenic delivery (AdVEC-m) is expected to provide an attractive vaccine strategy for cancer immunotherapy.


Assuntos
Adenoviridae/genética , Antígenos CD/imunologia , Caderinas/imunologia , Vacinas Anticâncer/imunologia , Portadores de Fármacos/química , Vetores Genéticos , Imunoterapia/métodos , Mananas/metabolismo , Adenoviridae/química , Animais , Anticorpos/sangue , Antígenos CD/genética , Western Blotting , Caderinas/genética , Vacinas Anticâncer/genética , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/prevenção & controle , Neoplasias/terapia , Neovascularização Patológica/imunologia , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia
20.
Med Oncol ; 27(4): 1156-63, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19949901

RESUMO

Colorectal cancer is one of the most common cancers. Survivin is strongly immunogenic in a fraction of colorectal cancer patients. The present study was designed to determine whether full-length mouse Survivin dominant-negative mutant SurvivinT34A has the antitumor activity in a murine colon carcinoma model. The complex of cationic liposome (DOTAP/Chol) to plasmid pORF9-mSurvivin T34A was administered intravenously in a mouse subcutaneous (S. C.) CT 26 tumor model. Apoptotic cells and anti-angiogenesis were evaluated by fluorescent in situ TUNEL assay and by immunohistochemistry with an antibody reactive to CD31, respectively. A 4 h 51Cr release assay was performed to determine Survivin-specific cytotoxicity. The adoptive transfer of CD8+ or CD4+ T-lymphocytes assay was to further explore the roles of immune cell subsets. We demonstrated the complex of cationic liposome (DOTAP/Chol) to plasmid pORF9--mSurvivin T34A when administered intravenously induced an efficient antitumor activity in a S. C. CT26 tumor model in mice. The main mechanism is involved in three aspects: triggering the apoptosis of tumor cells, inhibiting angiogenesis, and inducing Survivin-specific immune response. Our observations may have potential implications for the further exploration of the treatment of human colorectal cancer by intravenous delivery of dominant-negative mutant Survivin T34A.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/terapia , Terapia Genética , Proteínas Inibidoras de Apoptose/metabolismo , Neovascularização Patológica/prevenção & controle , Proteínas Repressoras/metabolismo , Transferência Adotiva , Alanina/genética , Animais , Apoptose , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Adesão Celular , Movimento Celular , Proliferação de Células , Neoplasias do Colo/imunologia , Feminino , Humanos , Técnicas Imunoenzimáticas , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/imunologia , Injeções Intravenosas , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Survivina , Treonina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA