1.Anti-tumor activity of wogonin, an extract from Scutellaria baicalensis, through regulating different signaling pathways.
Do Luong HUYNH ; Neelesh SHARMA ; Amit KUMAR SINGH ; Simrinder SINGH SODHI ; Jiao-Jiao ZHANG ; Raj Kumar MONGRE ; Mrinmoy GHOSH ; Nameun KIM ; Yang HO PARK ; Dong KEE JEONG
Chinese Journal of Natural Medicines (English Ed.) 2017;15(1):15-40
Wogonin is a plant flavonoid compound extracted from Scutellaria baicalensis (Huang-Qin or Chinese skullcap) and has been studied thoroughly by many researchers till date for its anti-viral, anti-oxidant, anti-cancerous and neuro-protective properties. Numerous experiments conducted in vitro and in vivo have demonstrated wogonin's excellent tumor inhibitory properties. The anti-cancer mechanism of wogonin has been ascribed to modulation of various cell signaling pathways, including serine-threonine kinase Akt (also known as protein kinase B) and AMP-activated protein kinase (AMPK) pathways, p53-dependent/independent apoptosis, and inhibition of telomerase activity. Furthermore, wogonin also decreases DNA adduct formation with a carcinogenic compound 2-Aminofluorene and inhibits growth of drug resistant malignant cells and their migration and metastasis, without any side effects. Recently, newly synthesized wogonin derivatives have been developed with impressive anti-tumor activity. This review is the succinct appraisal of the pertinent articles on the mechanisms of anti-tumor properties of wogonin. We also summarize the potential of wogonin and its derivatives used alone or as an adjunct therapy for cancer treatment. Furthermore, pharmacokinetics and side effects of wogonin and its analogues have also been discussed.
Animals
;
Antineoplastic Agents, Phytogenic
;
pharmacology
;
therapeutic use
;
DNA Adducts
;
metabolism
;
Drug Resistance, Neoplasm
;
drug effects
;
Drugs, Chinese Herbal
;
pharmacology
;
therapeutic use
;
Flavanones
;
pharmacology
;
therapeutic use
;
Humans
;
Neoplasms
;
drug therapy
;
metabolism
;
Phytotherapy
;
Scutellaria baicalensis
;
chemistry
;
Signal Transduction
;
drug effects
2.DNMT1 mediates chemosensitivity by reducing methylation of miRNA-20a promoter in glioma cells.
Daoyang ZHOU ; Yingfeng WAN ; Dajiang XIE ; Yirong WANG ; Junhua WEI ; Qingfeng YAN ; Peng LU ; Lianjie MO ; Jixi XIE ; Shuxu YANG ; Xuchen QI
Experimental & Molecular Medicine 2015;47(9):e182-
Although methyltransferase has been recognized as a major element that governs the epigenetic regulation of the genome during temozolomide (TMZ) chemotherapy in glioblastoma multiforme (GBM) patients, its regulatory effect on glioblastoma chemoresistance has not been well defined. This study investigated whether DNA methyltransferase (DNMT) expression was associated with TMZ sensitivity in glioma cells and elucidated the underlying mechanism. DNMT expression was analyzed by western blotting. miR-20a promoter methylation was evaluated by methylation-specific PCR. Cell viability and apoptosis were assessed using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and TdT-mediated dUTP-biotin nick end labeling assays, respectively. The results showed that compared with parental U251 cells, DNMT1 expression was downregulated, miR-20a promoter methylation was attenuated and miR-20a levels were elevated in TMZ-resistant U251 cells. Methyltransferase inhibition by 5-aza-2\'-deoxycytidine treatment reduced TMZ sensitivity in U251 cells. In U251/TM cells, DNMT1 expression was negatively correlated with miR-20a expression and positively correlated with TMZ sensitivity and leucine-rich repeats and immunoglobulin-like domains 1 expression; these effects were reversed by changes in miR-20a expression. DNMT1 overexpression induced an increase in U251/TM cell apoptosis that was inhibited by the miR-20a mimic, whereas DNMT1 silencing attenuated U251/TM cell apoptosis in a manner that was abrogated by miR-20a inhibitor treatment. Tumor growth of the U251/TM xenograft was inhibited by pcDNA-DNMT1 pretreatment and boosted by DNMT1-small hairpin RNA pretreatment. In summary, DNMT1 mediated chemosensitivity by reducing methylation of the microRNA-20a promoter in glioma cells.
Animals
;
Antineoplastic Agents, Alkylating/*pharmacology/therapeutic use
;
Apoptosis/drug effects
;
Brain/drug effects/metabolism/pathology
;
Brain Neoplasms/drug therapy/*genetics/pathology
;
DNA (Cytosine-5-)-Methyltransferase/antagonists & inhibitors/*genetics/metabolism
;
DNA Methylation
;
Dacarbazine/*analogs & derivatives/pharmacology/therapeutic use
;
Drug Resistance, Neoplasm
;
Female
;
Gene Expression Regulation, Neoplastic
;
Glioma/drug therapy/*genetics/pathology
;
Humans
;
Mice, Inbred C57BL
;
MicroRNAs/*genetics
;
Promoter Regions, Genetic
3.Effect of Peimine on ERCC1 mRNA and LRP Expressions of A549/DDP Multidrug Resistance Cell Line.
Xiao-yong TANG ; Ying-xue TANG ; Peng XU ; Hai-yan ZHOU ; Li HAN
Chinese Journal of Integrated Traditional and Western Medicine 2015;35(12):1490-1494
OBJECTIVETo explore the effect of peimine on excision repair cross-complementation 1 (ERCC1) mRNA and lung resistant protein (LRP) expressions in A549/cisplatin (DDP) multidrug resistance (MDR) cell line.
METHODSLung cancer A549/DDP cells were cultured in vitro.Cells at logarithmic growth phase were divided into 4 groups, i.e., the blank control group, the DDP group, the ligustrazine group (DDP+ligustrazine), the peimine group (DDP + peimine). After 48-h drug action, ERCC1 mRNA expression was detected by RT-PCR and LRP expression detected by cell immunofluorescence.
RESULTSThere was no statistical difference in expression levels of ERCC1 mRNA and LRP between the DDP group and the blank control group (P > 0.05). Compared with the DDP group, expression levels of ERCC1 mRNA and LRP obviously decreased in the ligustrazine group and the peimine group (P < 0.05). They were obviously lower in the peimine group than in the ligustrazine group (P < 0.05).
CONCLUSIONSPeimine could reverse MDR of A549/DDP cell line. Its mechanism might be associated with down-regulating ERCC1 mRNA and LRP expression levels.
Cell Line, Tumor ; Cevanes ; pharmacology ; Cisplatin ; DNA-Binding Proteins ; genetics ; Down-Regulation ; Drug Resistance, Multiple ; Drug Resistance, Neoplasm ; drug effects ; Endonucleases ; genetics ; Humans ; Low Density Lipoprotein Receptor-Related Protein-1 ; genetics ; Lung Neoplasms ; RNA, Messenger ; metabolism
4.DNA Hypomethylation-Mediated Overexpression of Carbonic Anhydrase 9 Induces an Aggressive Phenotype in Ovarian Cancer Cells.
Hye Youn SUNG ; Woong JU ; Jung Hyuck AHN
Yonsei Medical Journal 2014;55(6):1656-1663
PURPOSE: Both genetic and epigenetic alterations can lead to abnormal expression of metastasis-regulating genes in tumor cells. Recent studies suggest that aberrant epigenetic alterations, followed by differential gene expression, leads to an aggressive cancer cell phenotype. We examined epigenetically regulated genes that are involved in ovarian cancer metastasis. MATERIALS AND METHODS: We developed SK-OV-3 human ovarian carcinoma cell xenografts in mice. We compared the mRNA expression and DNA methylation profiles of metastatic tissues to those of the original SK-OV-3 cell line. RESULTS: Metastatic implants showed increased mRNA expression of the carbonic anhydrase 9 (CA9) gene and hypomethylation at CpG sites in the CA9 promoter. Treatment of wild-type SK-OV-3 cells with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine reduced methylation of the CA9 promoter and increased CA9 mRNA expression. Eight CpGs, which were located at positions -197, -74, -19, -6, +4, +13, +40, and +86, relative to the transcription start site, were hypomethylated in metastatic tumor implants, compared to that of wild-type SK-OV-3. Overexpression of CA9 induced an aggressive phenotype, including increased invasiveness and migration, in SK-OV-3 cells. CONCLUSION: Alterations in the DNA methylation profile of the CA9 promoter were correlated with a more aggressive phenotype in ovarian cancer cells.
Animals
;
Azacitidine/*analogs & derivatives/pharmacology
;
Carbonic Anhydrases/metabolism
;
*DNA Methylation
;
Female
;
Gene Expression Regulation, Neoplastic/*drug effects
;
Humans
;
Mice
;
Neoplasm Invasiveness/genetics
;
Neoplasm Metastasis/genetics/*pathology
;
Neoplasms, Experimental
;
Neoplasms, Glandular and Epithelial/genetics/*metabolism/pathology
;
Ovarian Neoplasms/genetics/*metabolism/pathology
;
Phenotype
;
Promoter Regions, Genetic
;
RNA, Messenger/metabolism
5.Effects of 5-Aza-dC on 5-Fu chemosensitivity by modulating TIP30 gene expression in human colorectal cancer cells.
Xiao-bing CHEN ; Bei-bei CHEN ; Jian LI ; Xin-feng WANG ; Yi-jie MA ; Su-xia LUO ; Hui-fang LÜ
Chinese Journal of Oncology 2013;35(11):824-827
OBJECTIVETo investigate the effect of 5-Aza-2'-deoxycytidine (5-Aza-dC) on TIP30 gene expression and the relationship between TIP30 expression and the sensitivity to 5-fluouracil (5-Fu) in colorectal cancer cells.
METHODSThe methylation profile of TIP30 gene in HCT116 colorectal cancer cells was determined by methylation-specific PCR. The levels of TIP30 mRNA and protein were determined by RT-PCR and Western blot after the 5-Aza-dC treatment. MTT assay was used to detect the chemosensitivity of HCT116 cells to 5-Fu.
RESULTSTIP30 gene displayed complete DNA methylation in the HCT116 cells without 5-Aza-dC pretreatment. After the 5-Aza-dC treatment for 3 days, only demethylating PCR amplification product was detected and TIP30 gene showed DNA demethylation. With the prolongation of the time of removal of 5-Aza-dC treatment, methylated and demethylated PCR amplification products were observed and TIP30 gene displayed both DNA methylation and DNA demethylation in the colorectal cancer cells. At the day 10 after removal of 5-Aza-dC, methylating PCR amplification product appeared and TIP30 gene showed DNA methylation. No expressions of TIP30 mRNA and protein were detected in the HCT116 cells untreated with 5-Aza-dC. After the treatment of 5-Aza-dC for 3 d and then removed the 5-Aza-dC, the expressions of TIP30 mRNA and protein were increased obviously. With the prolonged time after 5-Aza-dC removal, the expressions of TIP30 mRNA and protein decreased and reached the lowest level on day 10. The IC50 values of 5-Fu were 41.62, 33.17 and 4.96 µg/ml in the HCT116 cells pretreated with 5-Aza-dC, d0 and d10 with the drug removal after drug treatment for 3 d, respectively.
CONCLUSIONSThe results of this study show that the expression of TIP30 gene may be associated with its DNA methylation status and may affect the sensitivity of colorectal cancer cells to 5-Fu.
Acetyltransferases ; genetics ; metabolism ; Antimetabolites, Antineoplastic ; pharmacology ; Azacitidine ; analogs & derivatives ; pharmacology ; Cell Proliferation ; drug effects ; CpG Islands ; genetics ; DNA Methylation ; drug effects ; Drug Resistance, Neoplasm ; drug effects ; Fluorouracil ; pharmacology ; Gene Expression Regulation, Neoplastic ; HCT116 Cells ; Humans ; Inhibitory Concentration 50 ; RNA, Messenger ; metabolism ; Transcription Factors ; genetics ; metabolism
6.Suppression of the growth of subcutaneous transplanted human liver cancer and lung metastasis in nude mice treated by sorafenib combined with fluorouracil.
Hu-jia SHEN ; Yan-hong WANG ; Jian XU
Chinese Journal of Oncology 2013;35(2):98-102
OBJECTIVEThe aim of this study was to explore the inhibitory effect of sorafenib and 5-Fu on transplanted human liver cancer in nude mice, and to investigate the synergistic effect and mechanism between sorafenib and 5-Fu.
METHODSThe nude mouse model of human liver cancer was made by transplantation of human highly metastatic liver cancer cell line HCCLM3 cells, and the tumor-bearing nude mice were treated with sorafenib, 5-Fu or both, respectively, and mock-treated tumor-bearing nude mice as negative control. To assess the anti-tumor effect of sorafenib and the synergistic effect of sorafenib combined with 5-Fu by measuring the tumor weight and number of lung metastases. Moreover, the expressions of phosphorylated extracellular signal-regulated kinase (p-ERK), P-glycoprotein (P-gp) and topoisomerase 2-alpha (Topo IIa) in the nude mice were assayed by immunocytochemistry and Western blot.
RESULTSThe tumor weights and numbers of lung metastases were: (2.7 ± 0.825) g and 12.714 ± 6.317 in the negative control group, (0.933 ± 0.333) g and 4.333 ± 3.983 in the sorafenib group, (0.786 ± 0.212) g and 5.429 ± 4.315 in the Sorafenib + 5-Fu combination group, and (2.438 ± 0.793) g and 10.429 ± 6.241 in the 5-Fu group. Statistically, the tumor weights and numbers of lung metastases in the sorafenib group and combination group were significantly decreased, compared with that in the control group (P < 0.05). There was no significant difference in the tumor weight and number of lung metastases between the sorafenib group and the combination treatment group (P > 0.05). The expression levels of p-ERK, P-gp and Topo IIa proteins in the tumors after normalization were: negative control (0.017 ± 0.010, 0.085 ± 0.012, 0.103 ± 0.093), sorafenib group (0.010 ± 0.008, 0.044 ± 0.020, 0.020 ± 0.018), combination group (0.011 ± 0.007, 0.043 ± 0.023, 0.062 ± 0.026), and 5-Fu group (0.018 ± 0.009, 0.063 ± 0.032, 0.065 ± 0.034), respectively. Statistically, the expression of p-ERK, P-gp and Topo IIa in the Sorafenib group was significantly reduced compared with that of the control group (P < 0.05), and there was no significant difference in the expression of p-ERK, P-gp and Topo IIa between the sorafenib group and the combination treatment group (P > 0.05).
CONCLUSIONSSorafenib can inhibit not only the tumor growth and lung metastsis in the nude mouse models, but also reduce the expression of multidrug resistance proteins P-gp and Topo IIa as well. There is no significant advantage for the sorafenib + 5-Fu combination treatment than Sorafenib alone in inhibiting the expression of p-ERK, P-gp and Topo IIa.
ATP-Binding Cassette, Sub-Family B, Member 1 ; metabolism ; Animals ; Antimetabolites, Antineoplastic ; therapeutic use ; Antineoplastic Agents ; therapeutic use ; Cell Line, Tumor ; DNA Topoisomerases, Type II ; metabolism ; Drug Synergism ; Drug Therapy, Combination ; Extracellular Signal-Regulated MAP Kinases ; metabolism ; Fluorouracil ; therapeutic use ; Humans ; Liver Neoplasms ; drug therapy ; metabolism ; pathology ; Lung Neoplasms ; prevention & control ; secondary ; Male ; Mice ; Mice, Inbred BALB C ; Mice, Nude ; Neoplasm Transplantation ; Niacinamide ; analogs & derivatives ; therapeutic use ; Phenylurea Compounds ; therapeutic use ; Tumor Burden ; drug effects
7.Synthesis and structure-activity relationship of cycloberberine as anti-cancer agent.
Chong-Wen BI ; Cai-Xia ZHANG ; Yang-Biao LI ; Wu-Li ZHAO ; Rong-Guang SHAO ; Lin MEI ; Dan-Qing SONG
Acta Pharmaceutica Sinica 2013;48(12):1800-1806
A series of cycloberberine derivatives were designed, synthesized and evaluated for their anti-cancer activities in vitro. Among these analogs, compounds 6c, 6e and 6g showed strong inhibition on human HepG2 cells. They afforded a potent effect against DOX-resistant MCF-7 breast cells as well. The primary mechanism showed that cell cycle was blocked at G2/M phase of HepG2 cells treated with 6g using flow cytometry assay. It significantly inhibited the activity of DNA Top I at the concentration of 0.1 mg mL-1. Our results provided a basis for the development of this kind of compounds as novel anti-cancer agents.
Antineoplastic Agents
;
chemical synthesis
;
chemistry
;
pharmacology
;
Berberine
;
analogs & derivatives
;
chemical synthesis
;
chemistry
;
pharmacology
;
Cell Cycle
;
drug effects
;
Cell Proliferation
;
drug effects
;
DNA Topoisomerases, Type I
;
metabolism
;
Doxorubicin
;
pharmacology
;
Drug Resistance, Neoplasm
;
Hep G2 Cells
;
Humans
;
MCF-7 Cells
;
Molecular Structure
;
Structure-Activity Relationship
8.A facile, branched DNA assay to quantitatively measure glucocorticoid receptor auto-regulation in T-cell acute lymphoblastic leukemia.
Jason R SCHWARTZ ; Purvaba J SARVAIYA ; Lily E LEIVA ; Maria C VELEZ ; Tammuella C SINGLETON ; Lolie C YU ; Wayne V VEDECKIS
Chinese Journal of Cancer 2012;31(8):381-391
Glucocorticoid (GC) steroid hormones are used to treat acute lymphoblastic leukemia (ALL) because of their pro-apoptotic effects in hematopoietic cells. However, not all leukemia cells are sensitive to GC, and no assay to stratify patients is available. In the GC-sensitive T-cell ALL cell line CEM-C7, auto-up-regulation of RNA transcripts for the glucocorticoid receptor (GR) correlates with increased apoptotic response. This study aimed to determine if a facile assay of GR transcript levels might be promising for stratifying ALL patients into hormone-sensitive and hormone-resistant populations. The GR transcript profiles of various lymphoid cell lines and 4 bone marrow samples from patients with T-cell ALL were analyzed using both an optimized branched DNA (bDNA) assay and a real-time quantitative reverse transcription-polymerase chain reaction assay. There were significant correlations between both assay platforms when measuring total GR (exon 5/6) transcripts in various cell lines and patient samples, but not for a probe set that detects a specific, low abundance GR transcript (exon 1A3). Our results suggest that the bDNA platform is reproducible and precise when measuring total GR transcripts and, with further development, may ultimately offer a simple clinical assay to aid in the prediction of GC-sensitivity in ALL patients.
Adolescent
;
Antineoplastic Agents, Hormonal
;
pharmacology
;
Apoptosis
;
drug effects
;
Branched DNA Signal Amplification Assay
;
methods
;
Cell Line, Tumor
;
Child
;
Dexamethasone
;
pharmacology
;
Drug Resistance, Neoplasm
;
Exons
;
Glucocorticoids
;
pharmacology
;
Humans
;
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma
;
metabolism
;
pathology
;
Receptors, Glucocorticoid
;
genetics
;
metabolism
;
Reverse Transcriptase Polymerase Chain Reaction
;
methods
;
Transcription, Genetic
;
drug effects
;
Up-Regulation
9.Inhibitory effect of compound cantharides capsule on the proliferation of xenografts of human hepatocellular carcinoma HepG(2)215 in mice.
Jian-jun HAN ; Jin-ming YU ; Hui-yong WU ; Ji-bing LIU ; Bao SONG ; De-wen XUE
Chinese Journal of Oncology 2012;34(11):821-825
OBJECTIVETo investigate the inhibitory effect of compound cantharides capsules on the proliferation of xenografts of human hepatocellular carcinoma HepG(2215) in mice and their mechanism of action.
METHODSOne hundred healthy Balb/c mice (5-week old, male:female 1:1) were used in this study. Mouse models of human HepG(2215) hepatocarcinoma were established. The tumor-bearing mice were divided into five groups randomly. The control group A received daily intragastric administration of physiologic saline. The intervention groups B1, B2 and B3 were treated with compound cantharides capsule in a dose of 12.5 mg×kg(-1)×d(-1), 25 mg×kg(-1)×d(-1) and 37.5 mg×kg(-1)×d(-1), respectively, for 10 consecutive days. The group C had intraperitoneal injection of cyclophosphamide (25 mg×kg(-1)×d(-1)) for 10 consecutive days. The mice were sacrificed after the completion of administration. The tumors were taken out, the tumor volume was measured, the inhibitory rate of body weight was calculated, and the serum AFP concentration and the level of HBV DNA were determined. The survival of each group mice was analyzed. The levels of mRNA expression of apoptosis-related genes were assayed by quantitative RT-PCR. Apoptosis in the tumor cells was assayed with TUNEL staining. Flow cytometry was used to detect the levels of CD3(+), CD19(+), CD4(+) and CD8(+), and microvessel density (MVD) of the tumors was assessed by immunohistochemistry.
RESULTSAfter completion of the treatment, the inhibition rate of tumor growth of the groups B1, B2 and B3 was 29.8%, 38.7% and 48.1%, respectively, and that of the group C was 52.4%, with a significant difference among the groups (P < 0.05). The median survival time of the groups A, B1, B2, B3 and C was (30.0 ± 3.2) days, (49.0 ± 5.1) days, (50.0 ± 5.2) days, (57.5 ± 6.5) days and (49.0 ± 4.7) days, respectively. The median survival time of the group B3 was significantly longer than that of other groups (P < 0.05). The serum AFP level in the groups A, B1, B2, B3 and C was (492.7 ± 48.5) ng/ml, (281.2 ± 25.6) ng/ml, (194.3 ± 18.7) ng/ml, (170.1 ± 15.8) ng/ml and (138.7 ± 12.5) ng/ml, respectively, indicating that it was significantly inhibited in the group C. The inhibition rate of HBV DNA replication of the groups B1, B2, B3 and C was (46.0 ± 5.1)%, (65.5 ± 6.9)%, (81.3 ± 7.8)% and (19.5 ± 2.1)%, respectively, showing that compound cantharides capsules inhibited HBV DNA replication in a dose-dependent manner. The apoptosis rate of the groups A, B1, B2, B3 and C was (0.27 ± 0.03)%, (7.18 ± 2.12)%, (9.17 ± 2.42)%, (11.27 ± 3.03)% and (5.44 ± 2.45)%, respectively, and that of the group B3 was significantly higher than that of the groups A, B1, B2 and C (P < 0.05). The expression level of bax mRNA was significantly higher than that of the group C (P < 0.05). The drug could significantly decrease the bcl-2 mRNA expression level, more remarkably along with the increasing dose of cantharides, and it was significantly lower than that in the group C (P < 0.05). The levels of CD4(+), CD8(+), CD3(+) and CD19(+) were significantly higher than that in the groups A and C (P < 0.05). The value of MVD of the group B3 was significantly lower that that of groups A and C (P < 0.05).
CONCLUSIONCompound cantharides capsules may inhibit the replication of HBV DNA in HepG(2215) cells, inducing apoptosis in the tumor cells, enhancing the immune function to inhibit the growth of liver cancer cells in mice, and significantly prolong the median survival time of tumor-bearing mice.
Animals ; Antineoplastic Agents ; administration & dosage ; pharmacology ; Antiviral Agents ; administration & dosage ; pharmacology ; Apoptosis ; drug effects ; Cantharidin ; administration & dosage ; pharmacology ; Capsules ; DNA Replication ; DNA, Viral ; Drug Combinations ; Female ; Hep G2 Cells ; Hepatitis B virus ; Humans ; Male ; Mice ; Mice, Inbred BALB C ; Microvessels ; pathology ; Neoplasm Transplantation ; Proto-Oncogene Proteins c-bcl-2 ; genetics ; metabolism ; RNA, Messenger ; metabolism ; Tumor Burden ; drug effects ; Virus Replication ; Xenograft Model Antitumor Assays ; alpha-Fetoproteins ; metabolism ; bcl-2-Associated X Protein ; genetics ; metabolism
10.5-aza-2'-deoxycytidine-induced inhibition of CDH13 expression and its inhibitory effect on methylation status in human colon cancer cells in vitro and on growth of xenograft in nude mice.
Chinese Journal of Oncology 2012;34(1):6-10
OBJECTIVETo determine the inhibitory effect of 5-aza-2'-deoxycytidine (5-Aza-CdR) on the growth of human colon carcinoma cells and xenografts in nude mice, to observe its effect on CDH13 gene expression and methylation in the xenografts, and to explore the possible mechanisms.
METHODSHuman colon carcinoma cell line HCT116 cells were treated with 5-Aza-CdR, and the cell morphology was observe by phase contrast microscopy. The cell growth was assessed by MTT assay. A tumor-bearing mouse model was generated by subcutaneous inoculation of human colon carcinoma HCT116 cells into nude mice. The tumor growth in the nude mice was observed, the CDH13 gene expression and its methylation status in the tumors were detected using methylation specific PCR (MSP), RT-PCR, Western blotting and immunohistochemistry.
RESULTSAfter treatment with 5-Aza-CdR, the inhibition rate of the growth of cultured HCT116 cells was increased as the concentration was increasing. The growth of the xenografts in nude mice was significantly inhibited, and the methylated CDH13 gene was reactivated. After 4 weeks of 5-Aza-CdR treatment, no significant difference was found between the body weights of nude mice in the 5-Aza-CdR group [(18.06 ± 1.29) g] and control group [(17.07 ± 0.84) g], (P > 0.10), and the average volume of xenografts of the 5-Aza-CdR group was (907.00 ± 87.29) mm(3), significantly smaller than the (1370.93 ± 130.20) mm(3) in the control group (P < 0.005). No expression of CDH13 gene was found in the control group. The expression of CDH13 gene in the 5-Aza-CdR group was increased along with the increasing concentration of 5-Aza-CdR.
CONCLUSIONS5-Aza-CdR inhibits the growth of human colon cancer cells in culture and in nude mice, and induces the cancer cells to re-express CDH13 in nude mice. Its mechanism may be that demethylation of the methylated CDH13 promoter induced by 5-Aza-CdR restores CDH13 expression and thus inhibits the tumor growth in nude mice.
Animals ; Antimetabolites, Antineoplastic ; pharmacology ; Azacitidine ; analogs & derivatives ; pharmacology ; Cadherins ; genetics ; metabolism ; Cell Proliferation ; drug effects ; DNA Methylation ; HCT116 Cells ; Humans ; Male ; Mice ; Mice, Inbred BALB C ; Mice, Nude ; Neoplasm Transplantation ; RNA, Messenger ; metabolism ; Tumor Burden ; drug effects

Result Analysis
Print
Save
E-mail