Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Bioorg Chem ; 139: 106652, 2023 10.
Article in English | MEDLINE | ID: mdl-37390632

ABSTRACT

Primary liver cancer is one of the most common malignant cancers of the digestive system that lacks effective chemotherapeutic drugs in clinical settings. Camptothecin (CPT) and its derivatives have been approved for cancer treatment; however, their application is limited by their systemic toxicity. For lead optimization in new drug discovery stages, fluorination is an effective and robust approach to increase the bioavailability and optimize the pharmacokinetics of candidate compounds, thereby improving their efficacy. To obtain new and highly active CPT derivatives, we designed, synthesized, and evaluated two new fluorinated CPT derivatives, 9-fluorocamptothecin (A1) and 7-ethyl-9-fluorocamptothecin (A2), in this study. In vitro, A1 and A2 exhibited more robust anti-tumor activity than topotecan (TPT) in various cancer cells, particularly hepatocellular carcinoma (HCC) cells. In vivo, A1 and A2 exhibited greater anti-tumor activity than TPT in both AKT/Met induced primary HCC mouse models and implanted HepG2 cell xenografts. Acute toxicity tests revealed that A1 and A2 were not lethal and did not cause significant body weight loss at high doses. Moreover, A1 and A2 exhibited no significant toxicity in the mouse liver, heart, lung, spleen, kidney, and hematopoietic systems at therapeutic doses. Mechanistically, A1 and A2 blocked HCC cell proliferation by inhibiting the enzymatic activity of Topo I, subsequently inducing DNA damage, cell cycle arrest, and apoptosis. In summary, our results indicate that fluorination improves the anti-tumor activity of CPT while decreasing its toxicity and highlight the application potential of fluorination products A1 and A2 in clinical settings.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Camptothecin/pharmacology , Camptothecin/therapeutic use , Carcinoma, Hepatocellular/drug therapy , DNA Topoisomerases, Type I/metabolism , Liver Neoplasms/drug therapy , Topotecan/pharmacology , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Bioorg Chem ; 122: 105747, 2022 05.
Article in English | MEDLINE | ID: mdl-35338969

ABSTRACT

Based on the structural skeleton of natural products boeravinones, two types of 6H-chromeno[3,4-b]quinoline derivatives were designed and synthesized by nitrogen atom substitution strategy. Then, their cytotoxic activities were evaluated against six human tumor cell lines including HepG2 (hepatocellular carcinoma), A2780 (ovarian cancer), Hela (cervical cancer), HCT116 (colorectal cancer), SW1990 (pancreatic cancer), and MCF7 (breast cancer). The results showed that compounds ZML-8 and ZML-14 exhibited robust inhibitory activities against HepG2 cells with IC50 values of 0.58 and 1.94 µM, respectively. In addition, ZML-8 and ZML-14 showed higher selectivity against HepG2 and L-02 cells than Topotecan. Mechanistically, ZML-8 and ZML-14 not only induced cell cycle arrest in the G2/M phase and cell apoptosis, but also dose-dependently inhibited topoisomerase I activity and induced DNA damage in HepG2 cells. Molecular docking showed that ZML-8 and ZML-14 could interact with topoisomerase I-DNA complex with a similar binding mode to Topotecan. Inhibitory activities of these two compounds on topoisomerase I were then confirmed in both cell-free systems and in whole-cell lysates. Taken together, compounds ZML-8 and ZML-14 merit further development as a new generation of non-camptothecin topoisomerase I inhibitors for the treatment of cancer.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Antineoplastic Agents/chemistry , Apoptosis , Cell Line, Tumor , Cell Proliferation , Drug Design , Drug Screening Assays, Antitumor , Female , Humans , Molecular Docking Simulation , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors/pharmacology
3.
Bioorg Chem ; 114: 105065, 2021 09.
Article in English | MEDLINE | ID: mdl-34174631

ABSTRACT

29 novel 20(S)-aminophosphonate derivatives of camptothecin were synthesized via a FeCl3 - catalyzed one-pot reaction. All of these compounds displayed similar or superior cytotoxic activity in comparison with that of Irinotecan against Hep3B, MCF-7, A-549, MDA-MB-231, KB, and multidrug-resistant (MDR) KB-vin cell lines. Out of them, compound B07 exhibited significant cytotoxicity and 10-fold improvement in activity compared to Irinotecan. Mechanistically, B07 not only induced cell apoptosis and cell cycle arrest in Hep3B and MCF-7 cells, but also inhibited Topoisomerase I activity in the cell and cell-free system in a manner similar to that of Irinotecan. In both xenograft and primary HCC mouse models, B07 showed significant anti-tumor activity and was more potent than Irinotecan. Additionally, the acute toxicity assay showed that B07 had no apparent toxicity to the mouse liver, kidney, and hemopoietic system of the FVB/N mice. Therefore, these findings indicate that compound B07 could be a potential Topoisomerase I poison drug candidate for further clinical trial.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , Drug Design , Organophosphonates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Camptothecin/chemical synthesis , Camptothecin/chemistry , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Organophosphonates/chemical synthesis , Organophosphonates/chemistry , Structure-Activity Relationship
4.
Hepatogastroenterology ; 59(120): 2367-73, 2012.
Article in English | MEDLINE | ID: mdl-22773266

ABSTRACT

BACKGROUND/AIMS: MiR-93 was observed in various types of cancers. This study is to investigate a role of miR-93 in the carcinogenesis of HCC. METHODOLOGY: The expression of miR-93 in HepG2 cells and prima-ry human hepatocytes (PHHC) was measured by RT-PCR. HepG2 cells were transfected with miR-93 inhibitor or negative control. The cell proliferation was determined by using the CellTiter 96® Aqueous One Solution Cell Proliferation Assay kit. The migration and clonogenicity in vitro were measured by cell migration assay, colony formation analysis and anchorage-in-dependent growth assay. The apoptosis and cell cycle were detected by flow cytometry analysis. The mRNA and protein levels of transforming growth factor-beta type II receptor (TGFBR2) and integrin beta8 (ITGB8)were evaluated by RT-PCR and western blot analysis. RESULTS: MiR-93 was upregulated in HepG2 cells compared with PHHC and inhibition of miR-93 significantly suppressed HepG2 cell proliferation, migration and col-ony formation. The expressions of TGFBR2 and ITGB8 were upregulated when miR-93 was inhibited. CONCLUSIONS: Our results reveal an important contribution for miR-93 in hepatocarcinogenesis and suggest a role for TGFBR2 and ITGB8 dysregulation in this process. Thus,the use of synthetic inhibitor of miR-93 may prove to bea promising approach to liver cancer treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Proliferation , Liver Neoplasms/genetics , MicroRNAs/metabolism , Apoptosis/genetics , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Adhesion/genetics , Cell Cycle Checkpoints/genetics , Cell Movement/genetics , Down-Regulation , Flow Cytometry , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Stem Cell Assay
5.
Biochem Pharmacol ; 200: 115049, 2022 06.
Article in English | MEDLINE | ID: mdl-35469784

ABSTRACT

A series of new 7-ethyl-10-fluoro-20-O-(cinnamic acid ester)-camptothecin derivatives were synthesized and evaluated for cytotoxicity against four human tumor cell lines including HepG2 (hepatocellular carcinoma), SW480 (colorectal cancer), A2780 (ovarian cancer), and Hucct1 (intrahepatic cholangiocarcinoma). The results of cytotoxic activities in vitro showed that most of the camptothecin derivatives harbor promising cytotoxic activity against tested tumor cell lines. Among them, compound XJS-11 exhibited broad-spectrum inhibitory activities against HepG2, SW480, A2780, and Hucct1 cell lines with IC50 values of 0.03, 0.09, 0.22, and 0.32 µM, respectively. Further investigation demonstrated that compound XJS-11 exhibited more effective growth inhibition against a variety of human hepatoma cells (Sk-hep-1, Hep3B and Huh7) and lower cytotoxicity against immortalized normal human liver cell line L02 than the positive control topotecan. Especially, XJS-11 showed higher selective toxicity in two kinds of human hepatoma cells and immortalized normal human liver cell line (IC50(L-02)/IC50(HepG2) = 113.20; IC50(L-02)/IC50(Hep3B) = 85.60) than topotecan (IC50(L-02)/IC50(HepG2) = 9.45; IC50(L-02)/IC50(Hep3B) = 8.52). Mechanistically, XJS-11 induced cell cycle arrest and cell apoptosis in HepG2 and Hep3B cells by inhibiting Top I activity in a manner similar to that of topotecan. Meanwhile, XJS-11 could attenuate the tumor growth in both xenograft and primary HCC mouse models. In addition, the acute toxicity assay showed that XJS-11 did not cause lethality or significant body weight loss with a single intraperitoneal dose at 100 mg/kg or with an intraperitoneal dose at 25 mg/kg for 7 days. Moreover, unlike topotecan, XJS-11 had no apparent toxicity to the mouse liver, kidney, and hemopoietic system of the C57BL/6 mice. Taken together, XJS-11 merits further development as a new generation of the camptothecin-derived drug candidate.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Ovarian Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Camptothecin/pharmacology , Camptothecin/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cell Proliferation , Cinnamates , Drug Screening Assays, Antitumor , Esters , Female , Humans , Liver Neoplasms/drug therapy , Mice , Mice, Inbred C57BL , Topoisomerase I Inhibitors/pharmacology , Topoisomerase I Inhibitors/therapeutic use , Topotecan/pharmacology
6.
Life Sci ; 311(Pt A): 121105, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36272467

ABSTRACT

AIMS: In this study, we synthesized a 10-fluorine-substitution derivative of CPT (Camptothecin) YCJ100 and evaluated its antitumor activity and systemic toxicity. MATERIALS AND METHODS: Determination of in vitro antitumor activity and mechanism of YCJ100 by the MTT assay, Molecular docking, EdU staining, Cell cycle and apoptosis determination, Western blot analysis and Topoisomerase I activity assay. The antitumor effects of YCJ100 were evaluated in primary HCC (hepatocellular carcinoma), ICC (intrahepatic cholangiocarcinoma) mouse models, and pancreatic cancer xenograft models. KEY FINDINGS: YCJ100 showed superior cytotoxic activity compared to Topotecan in SW480, SW1990, Hep3B, HepG2, A549, A2780, HeLa, and QBC cells. YCJ100 blocked the cell cycle in the G2/M phase, inhibited cell proliferation and induced apoptosis in HepG2 and SW1990 cells. Mechanistically, YCJ100 inhibited topoisomerase I activity in both a cell-free system and a cellular system, similar to the mechanism of Topotecan. YCJ100 showed significant antitumor activity and was more potent than Topotecan in primary HCC and ICC mouse models, as well as a xenograft mouse model. Additionally, YCJ100 showed only minor toxicity to the mouse hematopoietic system, liver, and kidney. These findings indicate that YCJ100 has high antitumor activity and low systemic toxicity. SIGNIFICANCE: Our findings demonstrate that YCJ100, as a Topoisomerase I inhibitor, has in vitro and in vitro antitumor activity. This study provides a new lead compound worthy of further preclinical evaluation and potential clinical development.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Pancreatic Neoplasms , Topoisomerase I Inhibitors , Topotecan , Animals , Female , Humans , Mice , Antineoplastic Agents/pharmacology , Camptothecin , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Molecular Docking Simulation , Topotecan/pharmacology , Xenograft Model Antitumor Assays , Topoisomerase I Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy
7.
Curr Med Sci ; 40(2): 199-205, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32337681

ABSTRACT

Caspase recruitment domain containing protein 9 (CARD9) is an adaptor protein that plays a critical role in pattern recognition receptors (PRRs)-mediated activation of NF-?B and mitogen-activated protein kinase (MAPK). This elicits initiation of the pro-inflammatory cytokines and leads to inflammatory responses, which has been recognized as a critical contributor to chronic inflammation. Current researches demonstrate that CARD9 is strongly associated with metabolic diseases, such as obesity, insulin resistance, atherosclerosis and so on. In this review, we summarize CARD9 signaling pathway and the role of CARD9 in metabolic diseases.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Metabolic Diseases/metabolism , Gene Expression Regulation , Humans , MAP Kinase Signaling System , Receptors, Pattern Recognition/metabolism
8.
Curr Med Sci ; 40(3): 595, 2020 06.
Article in English | MEDLINE | ID: mdl-32681266

ABSTRACT

The article "The Role of CARD9 in Metabolic Diseases", written by Cheng TIAN, Ya-li TUO, Yi LU, Chuan-rui XU, Ming XIANG, was originally published electronically on the publisher's internet portal on May 2020 without open access. With the author(s)' decision to opt for Open Choice, the copyright of the article is changed to © The Author(s) 2020 and the article is forthwith distributed under a Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ ), which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.The original article has been corrected.

9.
Curr Med Sci ; 39(6): 929-937, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31845224

ABSTRACT

The molecular mechanisms underlying the development of intrahepatic cholangiocarcinoma (ICC) are not clear yet. In this study, we investigated the involvement of Notch1 in the development of ICC. The cDNA microarray analysis showed that Notch1 expression was higher in ICC tissues than in normal biliary epithelial cells. Stable transfection of Notch1 receptor intracellular domain (NICD1) by hydrodynamic tail vein injection induced ICC formation in mice. Western blotting confirmed that Notch1 signaling was activated in human ICC cell lines and mouse ICC tissues. Silencing Notch1 with specific short interfering RNA (siRNA) inhibited the proliferation of ICC cells. Flow cytometry and Western blotting indicated that apoptosis was induced in Notch1-silenced ICC cells compared with controls. Additionally, Notch1 silencing was associated with the inhibition of hairy and enhancer of split-1 (Hes1) and activation of the phosphatase and tensin homolog (PTEN)/p53 pathway. Taken together, these data suggest that Notch1 drives ICC formation and proliferation; downregulation of Notch1 induces apoptosis in ICC cells; Notch1 signaling may serve as a novel therapeutic target for the treatment of ICC.


Subject(s)
Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Oligonucleotide Array Sequence Analysis , Signal Transduction , Up-Regulation
10.
Oncotarget ; 7(52): 86675-86686, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27880727

ABSTRACT

MiR-375 is a tumor suppressor miRNA that is downregulated in hepatocellular carcinoma (HCC). However, due to the lack of effective delivery strategies, miR-375 replacement as a therapy for HCC has not been investigated. In the present study, we have developed a straightforward strategy to deliver miR-375 into HCC cells by assembling miR-375 mimics on the surface of AuNPs and forming AuNP-miR-375 nanoparticles. AuNP-miR-375 exhibits high cellular uptake and preserves miR-375's activities to suppress cellular proliferation, migration/invasion, and colony formation, and to induce apoptosis in HCC cells. Furthermore, AuNP-delivered miR-375 efficiently downregulated its target genes through RNA interference. In primary and xenograft tumor mouse models, AuNP-miR-375 showed high tumor uptake, therapeutic efficacy, and no apparent toxicity to the host mice. In conclusion, our findings indicate that AuNPs is a reliable strategy to deliver miR-375 into HCC cells and tissue, and that AuNP-miR-375 has the potential in the clinic for treatment of unresectable HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Gold/administration & dosage , Liver Neoplasms/drug therapy , Metal Nanoparticles/administration & dosage , MicroRNAs/administration & dosage , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , MicroRNAs/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
11.
World J Gastroenterol ; 11(46): 7368-73, 2005 Dec 14.
Article in English | MEDLINE | ID: mdl-16437645

ABSTRACT

AIM: To explore the possibility of repression of chloromycetin (Cm) acyl transferase by using external guided sequence (EGS) in order to converse the clinical E coli isolates from Cm- resistant to Cm- sensitive. METHODS: EGS directed against chloromycetin acetyl transferase gene (cat) was cloned to vector pEGFP-C1 which contains the kanamycin (Km) resistance gene. The recombinant plasmid pEGFP-C1+EGScat1+cat2 was constructed and the blank vector without EGS fragment was used as control plasmids. By using the CaCl(2) transformation method, the recombinant plasmids were introduced into the clinically isolated Cm resistant but Km sensitive E coli strains. Transformants were screened on LB agar plates containing Km. Extraction of plasmids and PCR were applied to identify the positive clones. The growth curve of EGS transformed bacteria cultured in broth with Cm resistance was determined by using spectrophotometer at A(600). Drug sensitivity was tested in solid culture containing Cm by using KB method. RESULTS: Transformation studies were carried out on 16 clinically isolated Cm-resistant (250 microg/mL of Cm) E coli strains by using pEGFP-C1-EGScat1cat2 recombinant plasmid. Transformants were screened on LB-agar plates containing Km after the transformation using EGS. Of the 16 tested strains, 4 strains were transformed successfully. Transformants with EGS plasmid showed growth inhibition when grown in liquid broth culture containing 200 microg/mL of Cm. In drug sensitivity test, these strains were sensitive to Cm on LB-agar plates containing 200 microg/mL of Cm. Extraction of plasmids and PCR amplification showed the existence of EGS plasmids in these four transformed strains. These results indicated that the Cat of the four clinical isolates had been suppressed and the four strains were converted to Cm sensitive ones. CONCLUSION: The EGS directed against Cat is able to inhibit the expression of Cat, and hence convert Cm-resistant bacteria to Cm-sensitive ones. Thus, the EGS has the capability of converting the phenotype of clinical drug-resistant isolates strains to drug-sensitive ones.


Subject(s)
Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol Resistance , Escherichia coli/drug effects , Escherichia coli/enzymology , Base Sequence , Chloramphenicol O-Acetyltransferase/antagonists & inhibitors , Chloramphenicol Resistance/genetics , DNA, Bacterial/genetics , Escherichia coli/genetics , Genes, Bacterial , Humans , In Vitro Techniques , Mutation , Phenotype , Transformation, Genetic
12.
J Huazhong Univ Sci Technolog Med Sci ; 35(5): 730-735, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26489630

ABSTRACT

Bmi1 is a member of the polycomb group family of proteins, and it drives the carcinogenesis of various cancers and governs the self-renewal of multiple types of stem cells. However, its role in the initiation and progression of bladder cancer is not clearly known. The present study aimed to investigate the function of Bmi1 in the development of bladder cancer. Bmi1 expression was detected in human bladder cancer tissues and their adjacent normal tissues (n=10) by immunohistochemistry, qRT-PCR and Western blotting, respectively. Bmi1 small interference RNA (siRNA) was synthesized and transfected into human bladder carcinoma cells (EJ) by lipofectamine 2000. The Bmil expression at mRNA and protein levels was measured in EJ cells transfected with Bmil siRNA (0, 80, 160 nmol/L) by qRT-PCR and Western blotting, respectively. Cell viability and Ki67 expression (a marker of cell proliferation) were determined in Bmi1 siRNA-transfected cells by CCK-8 assay and qRT-PCR, respectively. Cell cycle of transfected cells was flow-cytometrically determined. Immunofluorescence and Western blotting were used to detect the expression levels of cell cycle-associated proteins cyclin D1 and cyclin E in the cells. Pro-apoptotic proteins Bax and caspase 3 and anti-apoptotic protein Bcl-2 were detected by Western blotting as well. Additionally, xenograft tumor models were established by inoculation of EJ cells (infected with Bmil shRNA/pLKO.1 lentivirus or not) into nude mice. The tumor volumes were measured every other day for 14 days. The results showed that the Bmil expression was significantly increased in bladder tumor tissues when compared with that in normal tissues (P<0.05). Perturbation of Bmi1 expression by using siRNA could significantly inhibit the proliferation of EJ cells (P<0.05). Bmi1 siRNA-transfected EJ cells were accumulated in G1 phase and the expression levels of cyclin D1 and cyclin E were down-regulated. Bax and caspase-3 expression levels were significantly increased and Bcl-2 levels decreased after Bmi1 knockdown. Tumor volume was conspicuously reduced in mice injected with EJ cells with Bmi1 knockdown. Our findings indicate that Bmi1 is a potential driver oncogene of bladder cancer and it may become a potential treatment target for human bladder cancer.


Subject(s)
Carcinoma/therapy , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Polycomb Repressive Complex 1/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Urinary Bladder Neoplasms/therapy , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin E/antagonists & inhibitors , Cyclin E/genetics , Cyclin E/metabolism , Humans , Injections, Intralesional , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Burden , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Mol Biosyst ; 11(2): 532-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25424171

ABSTRACT

UNLABELLED: Emerging evidence indicates that microRNAs (miRNAs) are often dysregulated and play a fundamental role in hepatocellular carcinoma (HCC). However, the mechanism underlying miRNA dysregulation is still elusive. In the present study, we adopted an integrated analysis strategy combining data from genome-wide methylated DNA immunoprecipitation chip and miRNA expression microarray to study the regulation of DNA methylation on miRNA expression in HCC. We first characterized 864 differentially methylated regions (DMRs) located in 236 miRNA regions between cancerous and normal hepatocytes in HCC. We observed that the occurrence of miRNA DNA hypomethylation was more common than its hypermethylation while miRNA DNA hypermethylation was usually found in CpG islands. Then through correlation analysis between miRNA methylation and expression data, we identified 10 dysregulated miRNAs under the potential regulation of DNA methylation in HCC. Five of them (miR-148a, miR-375, miR-195, miR-497 and miR-378) were in hypermethylation and down-regulation status, while another five (miR-106b, miR-25, miR-93, miR-23a and miR-27a) were in hypomethylation and up-regulation status in HCC. Bioinformatics analysis showed that miR-148a may form a negative feedback loop with its targets DNMT1 and DNMT3B and the expression of the miR-195/497 cluster may be affected not only by their hypermethylated promoter region but also by their hypermethylated transcription factors NEUROG2 and DDIT3. CONCLUSION: our preliminary data and bioinformatics analysis suggest that DNA methylation plays an important and complex role in the regulation of miRNA expression in HCC, which may provide insights into the pathogenesis of HCC and thus may be used for diagnosis and intervention.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Cell Line, Tumor , Gene Regulatory Networks , Genes, Neoplasm , Hepatocytes/metabolism , Humans , Liver Neoplasms/genetics , MicroRNAs/metabolism , Transcription Factors/metabolism
14.
Oncol Rep ; 32(3): 1200-10, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24993451

ABSTRACT

Hepatocellular carcinoma (HCC) is a worldwide malignancy; however, there is a lack of effective targeted therapies. We and others have found that miR-221 is one of the most consistently overexpressed miRNAs in liver cancer. However, the roles of miR-221 in hepatocellular carcinogenesis are still not fully elucidated. In the present study, we used bioinformatics tools, gain- and loss-of-function methods to determine the roles of miR-221 in HCC. Bioinformatics analysis showed that miR-221 is a core miRNA which targets a large number of HCC-related genes and has formed many feed-forward regulatory loops combining transcription factors (TFs) to regulate HCC-related genes. Inhibition of miR-221 in liver cancer cells decreased cell proliferation, clonogenicity, migration/invasion and also induced G1 arrest and apoptosis. In addition, we demonstrated that miR-221 bound directly to the 3'-untranslated region of BMF, BBC3 and ANGPTL2, and inhibited the expression of BMF, BBC3 and ANGPTL2. In a mouse model, lentivirus­mediated miR-221 silencing could significantly suppress the growth of hepatoma xenografts in nude mice. In conclusion, we showed that miR-221 is a critical modulator in the HCC signaling pathway, and miR-221 silencing inhibits liver cancer malignant properties in vitro and in vivo, which may benefit the treatment for patients with unresectable HCC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Angiopoietins/genetics , Apoptosis Regulatory Proteins/genetics , Carcinoma, Hepatocellular/genetics , Computational Biology/methods , Liver Neoplasms/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , HeLa Cells , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation
15.
Mol Cancer Res ; 7(12): 1937-45, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19934271

ABSTRACT

Bmi1 is a polycomb group proto-oncogene that has been implicated in multiple tumor types. However, its role in hepatocellular carcinoma (HCC) development has not been well studied. In this article, we report that Bmi1 is overexpressed in human HCC samples. When Bmi1 expression is knocked down in human HCC cell lines, it significantly inhibits cell proliferation and perturbs cell cycle regulation. To investigate the role of Bmi1 in promoting liver cancer development in vivo, we stably expressed Bmi1 and/or an activated form of Ras (RasV12) in mouse liver. We found that while Bmi1 or RasV12 alone is not sufficient to promote liver cancer development, coexpression of Bmi1 and RasV12 promotes HCC formation in mice. Tumors induced by Bmi1/RasV12 resemble human HCC by deregulation of genes involved in cell proliferation, apoptosis, and angiogenesis. Intriguingly, we found no evidence that Bmi1 regulates Ink4A/Arf expression in both in vitro and in vivo systems of liver tumor development. In summary, our study shows that Bmi1 can cooperate with other oncogenic signals to promote hepatic carcinogenesis in vivo. Yet Bmi1 functions independent of Ink4A/Arf repression in liver cancer development.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation/genetics , Enzyme Activation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hepatocytes/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Up-Regulation/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL