Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Med Rep ; 24(5)2021 Nov.
Article in English | MEDLINE | ID: mdl-34490473

ABSTRACT

Since Professor Tu Youyou won the 2015 Nobel Prize in Physiology and Medicine for the discovery of artemisinin, which is used to treat malaria, increased attention has been paid to the extracts obtained from plants, in order to analyze their biological activities, particularly with regard to their antitumor activity. Therefore, the present study explored the biochemical properties of seven natural plant extracts on renal cell carcinoma (RCC). 786­O and OS­RC­2 cells were cultured and treated with different concentrations of the extracts. Then, cell viability, the IC50 value and proliferation was determined using a Cell Counting Kit­8 assay. Apoptosis and cell cycle distribution were evaluated via flow cytometry. The expression levels of proteins were assessed using western blotting, and cellular morphology was observed using a light microscope. The results showed that sophoricoside, aucubin, notoginsenoside R1 and ginsenoside Rg1 did not exhibit a cytotoxic effect on RCC cells, whereas ginsenoside Re and allicin exhibited a very slight inhibitory effect. Naringenin possessed the highest activity of the analyzed extracts. The IC50 values of naringenin on 786­O and OS­RC­2 cells were 8.91±0.33 and 7.78±2.65 µM, respectively. In addition, naringenin notably inhibited the proliferation of RCC cells by decreasing Ki67 expression, blocked cell cycle progression in the G2 phase by regulating expression of cell cycle proteins, and increased apoptosis by upregulating caspase­8 expression, downregulating Bcl­2 expression and altering the cellular morphology. Furthermore, naringenin inhibited cell proliferation and promoted apoptosis by upregulating the expression of PTEN at the protein level, downregulated the expression of PI3K and phosphorylated­(p­)AKT, but did not affect the expression of AKT, mTOR or p­mTOR. The seven plant extracts analyzed showed differing degrees of anti­RCC activity. Sophoricoside, aucubin, notoginsenoside R1 and ginsenoside Rg1 did not exhibit notable anti­RCC activity, whereas the effect of ginsenoside Re and allicin on RCC was considerably weak. However, naringenin showed potent anti­proliferative, apoptosis inducing and cell cycle arresting activity on RCC cells via regulation of the PTEN/PI3K/AKT signaling pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biological Products/pharmacology , Carcinoma, Renal Cell/drug therapy , Drugs, Chinese Herbal/pharmacology , Kidney Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism
2.
Iran J Public Health ; 47(5): 623-632, 2018 May.
Article in English | MEDLINE | ID: mdl-29922603

ABSTRACT

BACKGROUND: The aim of this study was to conduct a meta-analysis to estimate the association between the two SNPs and PCa risk. METHODS: Medline, Embase, Scopus, PubMed, Web of Science, Wan Fang Database and Chinese Zhi Wang Database were searched for the association of the two SNPs with susceptibility to PCa. The effect size was pooled by odds ratios (ORs) and 95% confidence intervals (95% CIs). RESULTS: Nine case-control studies, 5 on rs3787016 and 4 on rs2910164, were included. As regards rs3787016, an increased risk of PCa was identified in all genotype models (T versus C: OR = 1.18, 95% CI 1.11-1.25; CT versus CC: OR = 1.17, 95% CI 1.08-1.26; TT versus CC: OR = 1.41, 95% CI 1.22-1.63; TT + CT versus CC: OR = 1.20, 95% CI 1.12-1.30; TT versus CT + CC: OR = 1.32, 95% CI 1.15-1.52). However, no significant association was found between rs2910164 and PCa risk in any genetic models, in fact a trend of reduced risk could be seen (C versus G: OR = 0.91, 95% CI 0.79-1.05; GC versus GG: OR = 0.93, 95% CI 0.74-1.18; CC versus GG: OR = 0.70, 95% CI 0.47-1.02; CC + GC versus GG: OR = 0.90, 95% CI 0.73-1.12; CC versus GC + GG: OR = 0.78, 95% CI 0.56-1.08). Besides, in analysis of subgroups by source of controls, the decreased results were observed in studies using population-based controls. CONCLUSION: lncRNA POLR2E polymorphism rs3787016 is associated with a significantly increased risk of PCa, while a trend of reduced risk appears with mir-146a polymorphism rs2910164.

3.
Onco Targets Ther ; 11: 3267-3280, 2018.
Article in English | MEDLINE | ID: mdl-29910622

ABSTRACT

BACKGROUND: The vitamin D receptor (VDR) plays a key role in vitamin-mediated signaling pathway. Emerging evidence has suggested that the VDR polymorphism may contribute to the risk of prostate cancer (PCa). However, the existing results are not conclusive in Asian population. METHODS: We aim to evaluate the potential role of VDR polymorphisms on PCa of Asian population. PubMed, Scopus, Embase, Web of Science, Chinese National Knowledge Infrastructure, Wang Fang Data, and VIP Periodical were retrieved, and eligible studies (case-control or cohort study) meeting the inclusion criteria were evaluated through an updated meta-analysis using Stata13.0 software. RESULTS: A total of 1,363 cases and 2,101 controls obtained from 13 eligible publications were eventually included in this meta-analysis. Our results show that a significant association of VDR taq1 polymorphism with PCa risk, especially in the Japanese population. In the clinical stage-stratified analysis, the pooled results revealed no significant difference in genetic polymorphisms between the local stage and control groups, whereas there was increased frequency of T allele and TT genotype in the advanced tumor stage group compared with local tumor stage or control groups. Similarly, no significant difference was seen in Gleason <7 and control groups, but the T allele and TT genotype were significantly higher in the Gleason ≥7 group compared with Gleason <7 or control groups. CONCLUSION: The VDR TaqI polymorphism might be associated with PCa risk in Asian population, especially in the Japanese population. Also, PCa patients carrying the T allele or TT genotype were more likely to progress to advanced stage. These results suggest that VDR TaqI polymorphisms may be potential diagnostic biomarkers for PCa susceptibility.

4.
Acta Pharm ; 68(2): 211-222, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29702481

ABSTRACT

Bladder cancer (BCa) is one of the most common urinary cancers. The present study aims to investigate whether Paeoniflorin (Pae) can exert inhibitory effects on BCa. The results showed that Pae inhibited proliferation of human BCa cell lines in a concentration- and time-dependent manner. Pae and cisplatin (Cis) synergistically inhibited the growth of tumours in RT4-bearing mice. Pae treatment neutralized the body loss induced by Cis. Moreover, Pae induced apoptosis in RT4 cells and increased the activities of caspase3, caspase8 and caspase9. Western blotting and immunohistochemical analysis revealed that the phosphorylated signal transducer and activator of transcription-3 (p-STAT3) level were decreased in Pae-treated RT4 cells and Pae-treated tumour-bearing mice. Furthermore, STAT3 transcriptional target B-cell lymphoma-2 was decreased in Pae-treated RT4 cells. Interestingly, Pae prevented translocation of STAT3 to the nucleus in RT4 cells. Collectively, Pae inhibits the growth of BCa, at least in part, via a STAT3 pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Glucosides/pharmacology , Monoterpenes/pharmacology , STAT3 Transcription Factor/metabolism , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Dose-Response Relationship, Drug , Drug Synergism , Glucosides/administration & dosage , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Monoterpenes/administration & dosage , Phosphorylation/drug effects , Signal Transduction/drug effects , Time Factors , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
Mol Med Rep ; 15(6): 4191-4198, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28487942

ABSTRACT

Renal cell carcinoma (RCC) is the most frequently occurring malignancy of the kidney worldwide. Anti-angiogenic targeted therapies inhibit the progression of RCC, however, limited effects on the invasion or metastasis of tumor cells have been observed. Cyclic AMP responsive element­binding protein (CREB) is a serine/threonine kinase that has been implicated in the regulation of cell proliferation, apoptosis, cycle progression and metastasis, amongst others. Our previous research demonstrated that phosphorylated CREB (pCREB) was upregulated in human renal cancer cell lines and tissues, and decreased pCREB at the Ser133 site inhibited the growth and metastatic activity of OS­RC­2 cells. However, the role of CREB in RCC metastasis requires further investigation. Thus, the present study further investigated the role of CREB in RCC metastasis. The present study demonstrated that knockdown of CREB using small interfering RNA (siRNA) that targeted CREB (siCREB) significantly inhibited the migration and invasion of 786­O and OS­RC­2 cells, however, the opposite effect was observed in ACHN cells. In addition, knockdown of CREB suppressed the expression of matrix metallopeptidase (MMP)­2/9 and proteins associated with epithelial­mesenchymal transition (EMT) in 786­O and OS­RC­2 cells, and promoted expression in ACHN cells. Furthermore, the chromatin immunoprecipitation assay indicated that pCREB (Ser133) had a direct interaction with the fibronectin promoter, however, pCREB (Ser133) did not target the vimentin promoter in RCC. Therefore, the results of the present study indicate that CREB regulated metastatic RCC by mediating the expression of MMP­2/9 and EMT­associated proteins, however, CREB­mediated MMP­2/9 and EMT­associated protein expression may be induced by different pathways in different RCC cells.


Subject(s)
CREB-Binding Protein/metabolism , Carcinoma, Renal Cell/metabolism , Epithelial-Mesenchymal Transition/physiology , Kidney Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Metastasis/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , Vimentin/metabolism
6.
Mol Med Rep ; 15(6): 4273-4279, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28487984

ABSTRACT

Renal cell carcinoma (RCC) is the most common neoplasm of the kidney in adults, accounting for ~3% of adult malignancies. Understanding the underlying mechanism of RCC tumorigenesis is necessary to improve patient survival. The present study revealed that Taxol­induced microtubule (MT) polymerization causes cell cycle arrest and an increase in guanosine triphosphate­Ras homology gene family, member A (GTP­RhoA) protein expression. Disruption of Taxol­induced MT polymerization reversed GTP­RhoA expression and cell cycle arrest. The localization and redistribution of MTs and RhoA were consistent in cells with MT bundles and those without. Decreased GTP­RhoA had no marked effect on Taxol­induced MT bundling, however, it reduced the proportion of cells in G2/M phase. Taken together, Taxol­induced MT polymerization regulated the protein expression levels of GTP­RhoA and cell cycle arrest. However, the alteration in GTP­RhoA expression did not influence MT arrangement, suggesting that GTP­RhoA serves a pivotal role in Taxol­induced MT polymerization and cell cycle arrest in RCC.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Cell Cycle Checkpoints/drug effects , Kidney Neoplasms/drug therapy , Microtubules/drug effects , Paclitaxel/pharmacology , Polymerization/drug effects , rhoA GTP-Binding Protein/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints/drug effects , Guanosine Triphosphate/metabolism , Humans , Kidney Neoplasms/metabolism , Tubulin/metabolism
7.
Iran J Basic Med Sci ; 19(11): 1245-1250, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27917282

ABSTRACT

OBJECTIVES: Renal cell carcinoma (RCC) is insensitive to conventional chemotherapy. Ginkgetin effectively treats several carcinoma cells. However, little is known about effects of Ginkgetin on RCC. In the present study, using 786-O cells, we evaluate whether Ginkgetin exerts anticancer effects against RCC. MATERIALS AND METHODS: 786-O cells suspended in the medium containing Ginkgetin were cultured for 24 hr to 72 hr, and then MTT assay was used to study cytotoxic effect of Ginkgetin. Apoptosis in 786-O was measured by an FITC Annexin apoptosis detection kit. Protein expression was detected by Western blotting. 786-O cells with active Janus kinase 2 (JAK2)-Signal transducer and activator of transcription 3 (STAT3) were prepared by stimulant of interleukin-6 (IL-6), whereas 786-O cells with deactivated STAT3 were produced by small interfering RNA (siRNA) STAT3. RESULTS: Ginkgetin suppressed the growth of 786-O in dose and time-dependent manners with IC50 values of 7.23 µM. Ginkgetin induced apoptosis of 786-O cells and increased the levels of caspase-8, caspase-9, and caspase-3. Additionally, Ginkgetin treated 786-O cells showed decreased levels of JAK2 and phosphorylated-STAT3 whether or not IL-6 was pretreated. Interestingly, pretreatment of siRNA STAT3 exerted inhibitory effects on the growth of 786-O cells, and the observation could be further reinforced after the Ginkgetin treatment. CONCLUSION: Our results indicate Ginkgetin possesses obvious inhibitory effects on the proliferation of 786-O, and this effect is probably due to its inhibition of JAK2/STAT3 pathway. Our findings imply Ginkgetin is a potential therapeutic medicine for RCC.

8.
Mol Med Rep ; 14(6): 5614-5618, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27878266

ABSTRACT

The present study aimed to investigate the effects of cantharidin on cell cycle distribution, the induction of apoptosis, and Notch1 and Jagged1 expression in ACHN and Caki­1 renal cancer cells. Cell viability assay, flow cytometry, cell cycle and western blot analyses were performed for ACHN and Caki­1 cells. Immunohistochemistry was used to analyze the expression of Notch1 and Jagged1 in RCC tissues The results demonstrated that treatment with cantharidin exerted a dose­ and time­dependent effect on cell viability, apoptosis induction and G2/M phase cell cycle arrest. Exposure of ACHN and Caki­1 cells to 20 µM cantharidin reduced cell viability to 26 and 32% respectively, after 48 h. In addition, treatment with cantharidin enhanced the number of ACHN and Caki­1 cells in G2/M phase to 54.62 and 51.88% respectively, as compared with 17.16 and 16.53% in the control groups. In the ACHN and Caki­1 cells, treatment with cantharidin induced a marked increase in the proportion of apoptotic cells after 48 h. Furthermore, cantharidin enhanced the percentage ACHN and Caki­1 apoptotic cells to 57.23 and 62.34% respectively, as compared with 2.27 and 3.06% in the control groups. Detection of Notch1 and Jagged1 expression demonstrated that levels were significantly increased in carcinoma tissues. Conversely, cantharidin exhibited an inhibitory effect on Notch1 and Jagged1 expression after 48 h. Therefore, treatment with cantharidin may exert a promising effect on the inhibition of renal cancer, and may be of therapeutic importance for the treatment of renal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cantharidin/pharmacology , Enzyme Inhibitors/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Immunohistochemistry , Jagged-1 Protein/metabolism , Kidney Neoplasms/metabolism , Receptor, Notch1/metabolism , Signal Transduction/drug effects
9.
BMC Urol ; 16(1): 52, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27578166

ABSTRACT

BACKGROUND: Methylation of the tumor suppressor gene H-cadherin (CDH13) has been reported in many cancers. However, the clinical effect of the CDH13 methylation status of patients with bladder cancer remains to be clarified. METHODS: A systematic literature search was performed to identify eligible studies in the PubMed, Embase, EBSCO, CKNI and Wanfang databases. The pooled odds ratio (OR) and the corresponding 95 % confidence interval (95 % CI) was calculated and summarized. RESULTS: Nine eligible studies were included in the present meta-analysis consisting of a total of 1017 bladder cancer patients and 265 non-tumor controls. A significant association was found between CDH13 methylation levels and bladder cancer (OR = 21.71, P < 0.001). The results of subgroup analyses based on sample type suggested that CDH13 methylation was significantly associated with bladder cancer risk in both the tissue and the urine (OR = 53.94, P < 0.001; OR = 7.71, P < 0.001; respectively). A subgroup analysis based on ethnic population showed that the OR value of methylated CDH13 was higher in Asians than in Caucasians (OR = 35.18, P < 0.001; OR = 8.86, P < 0.001; respectively). The relationships between CDH13 methylation and clinicopathological features were also analyzed. A significant association was not observed between CDH13 methylation status and gender (P = 0.053). Our results revealed that CDH13 methylation was significantly associated with high-grade bladder cancer, multiple bladder cancer and muscle invasive bladder cancer (OR = 2.22, P < 0.001; OR = 1.45, P = 0.032; OR = 3.42, P < 0.001; respectively). CONCLUSION: Our study indicates that CDH13 methylation may play an important role in the carcinogenesis, development and progression of bladder cancer. In addition, CDH13 methylation has the potential to be a useful biomarker for bladder cancer screening in urine samples and to be a prognostic biomarker in the clinic.


Subject(s)
Biomarkers, Tumor/metabolism , Cadherins/metabolism , Urinary Bladder Neoplasms/metabolism , Cadherins/genetics , Humans , Methylation , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...