Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 17(3): 794-805, 2019 03.
Article in English | MEDLINE | ID: mdl-30610105

ABSTRACT

Methyl 2-trifluoromethyl-3,11-dioxo-18ß-olean-1,12-dien-3-oate (CF3DODA-Me) is derived synthetically from glycyrrhetinic acid, a major component of licorice, and this compound induced reactive oxygen species (ROS) in RD and Rh30 rhabdomyosarcoma (RMS) cells. CF3DODA-Me also inhibited growth and invasion and induced apoptosis in RMS cells, and these responses were attenuated after cotreatment with the antioxidant glutathione, demonstrating the effective anticancer activity of ROS in RMS. CF3DODA-Me also downregulated expression of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 and prooncogenic Sp-regulated genes including PAX3-FOXO1 (in Rh30 cells). The mechanism of CF3DODA-Me-induced Sp-downregulation involved ROS-dependent repression of c-Myc and cMyc-regulated miR-27a and miR-17/20a, and this resulted in induction of the miRNA-regulated Sp repressors ZBTB4, ZBTB10, and ZBTB34. The cell and tumor growth effects of CF3DODA-Me further emphasize the sensitivity of RMS cells to ROS inducers and their potential clinical applications for treating this deadly disease. IMPLICATIONS: CF3DODA-Me and HDAC inhibitors that induce ROS-dependent Sp downregulation could be developed for clinical applications in treating rhabdomyosarcoma.


Subject(s)
Reactive Oxygen Species/metabolism , Rhabdomyosarcoma/drug therapy , Sp Transcription Factors/genetics , Triterpenes/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Rhabdomyosarcoma/pathology , Transfection , Triterpenes/pharmacology
2.
PLoS One ; 13(2): e0192264, 2018.
Article in English | MEDLINE | ID: mdl-29389953

ABSTRACT

Metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1) is a long non-coding RNA (lncRNA) that is a negative prognostic factor for patients with pancreatic cancer and several other tumors. In this study, we show that knockdown of MALAT-1 in Panc1 and other pancreatic cancer cell lines decreases cell proliferation, survival and migration. We previously observed similar results for the lncRNAs HOTTIP and HOTAIR in Panc1 cells; however, RNAseq comparison of genes regulated by MALAT-1 shows minimal overlap with HOTTIP/HOTAIR-regulated genes. Analysis of changes in gene expression after MALAT-1 knockdown shows that this lncRNA represses several tumor suppressor-like genes including N-myc downregulated gene-1 (NDRG-1), a tumor suppressor in pancreatic cancer that is also corepressed by EZH2 (a PRC2 complex member). We also observed that Specificity proteins Sp1, Sp3 and Sp4 are overexpressed in Panc1 cells and Sp knockdown or treatment with small molecules that decrease Sp proteins expression also decrease MALAT-1 expression. We also generated Kras-overexpressing p53L/L;LSL-KrasG12DL/+;p48Cre+/- (p53L/L/KrasG12D) and p53L/+;LSLKrasG12DL/+;p48Cre+/- (p53L/+/KrasG12D) mice which are p53 homo- and heterozygous, respectively. These mice rapidly develop pancreatic ductal adenocarcinoma-like tumors and were crossed with MALAT-1-/- mice. We observed that the loss of one or two MALAT-1 alleles in these Ras overexpressing mice does not significantly affect the time to death; however, the loss of MALAT-1 in the p53-/+ (heterozygote) mice slightly increases their lifespan.


Subject(s)
Biomarkers, Tumor/genetics , Pancreatic Neoplasms/pathology , RNA, Long Noncoding/genetics , Animals , Cell Line, Tumor , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Mice, Transgenic , Pancreatic Neoplasms/genetics
3.
J Biol Chem ; 291(53): 27122-27133, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27875298

ABSTRACT

The antineoplastic agent benzyl isothiocyanate (BITC) acts by targeting multiple pro-oncogenic pathways/genes, including signal transducer and activator of transcription 3 (STAT3); however, the mechanism of action is not well known. As reported previously, BITC induced reactive oxygen species (ROS) in Panc1, MiaPaCa2, and L3.6pL pancreatic cancer cells. This was accompanied by induction of apoptosis and inhibition of cell growth and migration, and these responses were attenuated in cells cotreated with BITC plus glutathione (GSH). BITC also decreased expression of specificity proteins (Sp) Sp1, Sp3, and Sp4 transcription factors (TFs) and several pro-oncogenic Sp-regulated genes, including STAT3 and phospho-STAT3 (pSTAT3), and GSH attenuated these responses. Knockdown of Sp TFs by RNA interference also decreased STAT3/pSTAT3 expression. BITC-induced ROS activated a cascade of events that included down-regulation of c-Myc, and it was also demonstrated that c-Myc knockdown decreased expression of Sp TFs and STAT3 These results demonstrate that in pancreatic cancer cells, STAT3 is an Sp-regulated gene that can be targeted by BITC and other ROS inducers, thereby identifying a novel therapeutic approach for targeting STAT3.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Isothiocyanates/pharmacology , Pancreatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Sp Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/therapeutic use , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Oncotarget ; 6(13): 10840-52, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25912306

ABSTRACT

HOTTIP is a long non-coding RNA (lncRNA) transcribed from the 5' tip of the HOXA locus and is associated with the polycomb repressor complex 2 (PRC2) and WD repeat containing protein 5 (WDR5)/mixed lineage leukemia 1 (MLL1) chromatin modifying complexes. HOTTIP is expressed in pancreatic cancer cell lines and knockdown of HOTTIP by RNA interference (siHOTTIP) in Panc1 pancreatic cancer cells decreased proliferation, induced apoptosis and decreased migration. In Panc1 cells transfected with siHOTTIP, there was a decrease in expression of 757 genes and increased expression of 514 genes, and a limited gene analysis indicated that HOTTIP regulation of genes is complex. For example, Aurora kinase A, an important regulator of cell growth, is coregulated by MLL and not WDR5 and, in contrast to previous studies in liver cancer cells, HOTTIP does not regulate HOXA13 but plays a role in regulation of several other HOX genes including HOXA10, HOXB2, HOXA11, HOXA9 and HOXA1. Although HOTTIP and the HOX-associated lncRNA HOTAIR have similar pro-oncogenic functions, they regulate strikingly different sets of genes in Panc1 cells and in pancreatic tumors.


Subject(s)
Cell Movement , Cell Proliferation , Pancreatic Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Animals , Apoptosis , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Cell Line, Tumor , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice, Nude , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , RNA Interference , RNA, Long Noncoding/genetics , RNAi Therapeutics , Signal Transduction , Time Factors , Transfection , Xenograft Model Antitumor Assays
5.
Neoplasia ; 16(12): 1059-69, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25499219

ABSTRACT

ZBTB4 is a transcriptional repressor and examination of publically-available microarray data sets demonstrated an inverse relationship in the prognostic value and expression of ZBTB4 and the histone methyltransferase EZH2 in tumors from breast cancer patients. The possibility of functional interactions between EZH2 and ZBTB4 was investigated in breast cancer cells and the results showed that EZH2 is directly suppressed by ZBTB4 which in turn is regulated (suppressed) by miR-106b and other paralogues from the miR-17-92, miR-106b-25 and miR-106a-363 clusters that are highly expressed in breast and other tumors. ZBTB4 also acts a suppressor of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4, and RNA interference studies show that Sp proteins are required for EZH2 expression. The prediction analysis results from breast cancer patient array data sets confirm an association of Sp1-dependent EZH2 gene signature with decreased survival of breast cancer patients. Disruption of oncogenic miR-ZBTB4 signaling axis by anticancer agent such as betulinic acid that induce down-regulation of Sp proteins in breast cancer cells resulted in inhibition of tumor growth and colonization of breast cancer cells in a mouse model. Thus, EZH2 is reciprocally regulated by a novel signaling network consisting of Sp proteins, oncogenic miRs and ZBTB4, and modulation of this gene network is a novel therapeutic approach for treatment of breast cancer and possibly other cancers.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/physiology , MicroRNAs/genetics , Polycomb Repressive Complex 2/genetics , Repressor Proteins/genetics , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Blotting, Western , Breast Neoplasms/diagnosis , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Pentacyclic Triterpenes , Real-Time Polymerase Chain Reaction , Sp Transcription Factors/genetics , Transplantation, Heterologous , Triterpenes/therapeutic use , Tumor Cells, Cultured , Betulinic Acid
6.
Expert Opin Ther Targets ; 18(7): 759-69, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24793594

ABSTRACT

INTRODUCTION: Specificity protein (Sp) transcription factors (TFs) are members of the Sp/Kruppel-like factor family, and Sp proteins play an important role in embryonic and early postnatal development. Sp1 has been the most extensively investigated member of this family, and expression of this protein decreases with age, whereas Sp1 and other family members (Sp3 and Sp4) are highly expressed in tumors and cancer cell lines. AREA COVERED: The prognostic significance of Sp1 in cancer patients and the functional pro-oncogenic activities of Sp1, Sp3 and Sp4 in cancer cell lines are summarized. Several different approaches have been used to target downregulation of Sp TFs and Sp-regulated genes, and this includes identification of different structural classes of antineoplastic agents including NSAIDs, natural products and their synthetic analogs and several well-characterized drugs including arsenic trioxide, aspirin and metformin. The multiple pathways involved in drug-induced Sp downregulation are also discussed. EXPERT OPINION: The recognition by the scientific and clinical community that experimental and clinically used antineoplastic agents downregulate Sp1, Sp3 and Sp4, and pro-oncogenic Sp-regulated genes will facilitate future clinical applications for individual drug and drug combination therapies that take advantage of their unusual effects.


Subject(s)
Neoplasms/metabolism , Sp1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Down-Regulation , Humans , Neoplasms/drug therapy
7.
Mol Cell Biol ; 34(13): 2382-95, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24732804

ABSTRACT

Reactive oxygen species (ROS)-inducing anticancer agents such as phenethylisothiocyanate (PEITC) activate stress pathways for killing cancer cells. Here we demonstrate that PEITC-induced ROS decreased expression of microRNA 27a (miR-27a)/miR-20a:miR-17-5p and induced miR-regulated ZBTB10/ZBTB4 and ZBTB34 transcriptional repressors, which, in turn, downregulate specificity protein (Sp) transcription factors (TFs) Sp1, Sp3, and Sp4 in pancreatic cancer cells. Decreased expression of miR-27a/miR-20a:miR-17-5p by PEITC-induced ROS is a key step in triggering the miR-ZBTB Sp cascade leading to downregulation of Sp TFs, and this is due to ROS-dependent epigenetic effects associated with genome-wide shifts in repressor complexes, resulting in decreased expression of Myc and the Myc-regulated miRs. Knockdown of Sp1 alone by RNA interference also induced apoptosis and decreased pancreatic cancer cell growth and invasion, indicating that downregulation of Sp transcription factors is an important common mechanism of action for PEITC and other ROS-inducing anticancer agents.


Subject(s)
Anticarcinogenic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Isothiocyanates/pharmacology , Reactive Oxygen Species/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , MicroRNAs/biosynthesis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Small Interfering , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sp1 Transcription Factor/biosynthesis , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/biosynthesis , Sp4 Transcription Factor/biosynthesis , Xenograft Model Antitumor Assays
8.
Carcinogenesis ; 34(12): 2870-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23803693

ABSTRACT

Metformin is a widely used antidiabetic drug, and epidemiology studies for pancreatic and other cancers indicate that metformin exhibits both chemopreventive and chemotherapeutic activities. Several metformin-induced responses and genes are similar to those observed after knockdown of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 by RNA interference, and we hypothesized that the mechanism of action of metformin in pancreatic cancer cells was due, in part, to downregulation of Sp transcription factors. Treatment of Panc1, L3.6pL and Panc28 pancreatic cancer cells with metformin downregulated Sp1, Sp3 and Sp4 proteins and several pro-oncogenic Sp-regulated genes including bcl-2, survivin, cyclin D1, vascular endothelial growth factor and its receptor, and fatty acid synthase. Metformin induced proteasome-dependent degradation of Sps in L3.6pL and Panc28 cells, whereas in Panc1 cells metformin decreased microRNA-27a and induced the Sp repressor, ZBTB10, and disruption of miR-27a:ZBTB10 by metformin was phosphatase dependent. Metformin also inhibited pancreatic tumor growth and downregulated Sp1, Sp3 and Sp4 in tumors in an orthotopic model where L3.6pL cells were injected directly into the pancreas. The results demonstrate for the first time that the anticancer activities of metformin are also due, in part, to downregulation of Sp transcription factors and Sp-regulated genes.


Subject(s)
Cell Proliferation/drug effects , Down-Regulation/drug effects , Metformin/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Sp Transcription Factors/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Pancreatic Neoplasms/metabolism , Sp Transcription Factors/metabolism
9.
Mol Nutr Food Res ; 57(9): 1638-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23471840

ABSTRACT

SCOPE: Mechanisms involving the curcuminoids effects in decreasing the prooncogenic specificity protein (Sp) transcription factors, and Sp-regulated genes in SW-480 colon cancer cells and how the multidrug resistance protein (MDR1) inhibition is mediated by Sp suppression. METHODS AND RESULTS: HT-29 and SW-480 colon cancer and normal CCD-18Co colon fibroblast cells were treated with curcuminoids previously analyzed by HPLC. Gene and protein expression regulation were assessed by RT-PCR, transfections with expression constructs, and Western blots. Curcuminoids (2.5-10 µg/mL) suppressed preferentially the growth of SW-480 and HT-29 compared to CCD-18Co cells and enhanced the anticancer activity of the chemotherapeutic drug 5-fluorouracil due to the suppression of MDR1. Additionally, Sp1, Sp3, and Sp4 and Sp-regulated genes were downregulated by curcuminoids in SW-480 and this was accompanied by suppression of microRNA-27a (miR-27a) and induction of ZBTB10, an mRNA target of miR-27a and a transcriptional repressor of Sp expression. This mechanism was mediated by the induction of ROS. RNA-interference and transfection with ZBTB10-expression plasmid demonstrated that MDR1 was regulated by Sp1 and Sp3 and the disruption of the miR-27a-ZBTB10-Sp axis. CONCLUSION: Colon cancer treatment with curcuminoids will enhance the therapeutic effects of drugs in patients who have developed drug resistance.


Subject(s)
Colonic Neoplasms/metabolism , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Reactive Oxygen Species/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , HT29 Cells , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Plasmids/genetics , RNA Interference/drug effects , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Sp Transcription Factors/antagonists & inhibitors , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Transfection
10.
Int J Cancer ; 132(4): 795-806, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-22815231

ABSTRACT

Specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 are highly expressed in rhabdomyosarcoma (RMS) cells. In tissue arrays of RMS tumor cores from 71 patients, 80% of RMS patients expressed high levels of Sp1 protein, whereas low expression of Sp1 was detected in normal muscle tissue. The non-steroidal anti-inflammatory drug (NSAID) tolfenamic acid (TA) inhibited growth and migration of RD and RH30 RMS cell lines and also inhibited tumor growth in vivo using a mouse xenograft (RH30 cells) model. The effects of TA were accompanied by downregulation of Sp1, Sp3, Sp4 and Sp-regulated genes in RMS cells and tumors, and the role of Sp protein downregulation in mediating inhibition of RD and RH30 cell growth and migration was confirmed by individual and combined knockdown of Sp1, Sp3 and Sp4 proteins by RNA interference. TA treatment and Sp knockdown in RD and RH30 cells also showed that four genes that are emerging as individual drug targets for treating RMS, namely c-MET, insulin-like growth factor receptor (IGFR), PDGFRα and CXCR4, are also Sp-regulated genes. These results suggest that NSAIDs such as TA may have potential clinical efficacy in drug combinations for treating RMS patients.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/metabolism , Sp Transcription Factors/metabolism , ortho-Aminobenzoates/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation , Female , Humans , Mice , Mice, Nude , Muscles/metabolism , Proto-Oncogene Proteins c-met/genetics , RNA Interference , RNA, Small Interfering , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptors, CXCR4/genetics , Receptors, Somatomedin/genetics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Sp Transcription Factors/genetics , Xenograft Model Antitumor Assays
11.
PLoS One ; 7(10): e48208, 2012.
Article in English | MEDLINE | ID: mdl-23110215

ABSTRACT

Acetylsalicylic acid (aspirin) is highly effective for treating colon cancer patients postdiagnosis; however, the mechanisms of action of aspirin in colon cancer are not well defined. Aspirin and its major metabolite sodium salicylate induced apoptosis and decreased colon cancer cell growth and the sodium salt of aspirin also inhibited tumor growth in an athymic nude mouse xenograft model. Colon cancer cell growth inhibition was accompanied by downregulation of Sp1, Sp3 and Sp4 proteins and decreased expression of Sp-regulated gene products including bcl-2, survivin, VEGF, VEGFR1, cyclin D1, c-MET and p65 (NFκB). Moreover, we also showed by RNA interference that ß-catenin, an important target of aspirin in some studies, is an Sp-regulated gene. Aspirin induced nuclear caspase-dependent cleavage of Sp1, Sp3 and Sp4 proteins and this response was related to sequestration of zinc ions since addition of zinc sulfate blocked aspirin-mediated apoptosis and repression of Sp proteins. The results demonstrate an important underlying mechanism of action of aspirin as an anticancer agent and, based on the rapid metabolism of aspirin to salicylate in humans and the high salicylate/aspirin ratios in serum, it is likely that the anticancer activity of aspirin is also due to the salicylate metabolite.


Subject(s)
Aspirin/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Sp Transcription Factors/metabolism , Animals , Aspirin/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Sp Transcription Factors/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/genetics , Sp4 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
12.
Mol Cancer Ther ; 11(7): 1421-31, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22553354

ABSTRACT

Treatment of ErbB2-overexpressing BT474 and MDA-MB-453 breast cancer cells with 1 to 10 µmol/L betulinic acid inhibited cell growth, induced apoptosis, downregulated specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and decreased expression of ErbB2. Individual or combined knockdown of Sp1, Sp3, Sp4 by RNA interference also decreased expression of ErbB2 and this response was because of repression of YY1, an Sp-regulated gene. Betulinic acid-dependent repression of Sp1, Sp3, Sp4, and Sp-regulated genes was due, in part, to induction of the Sp repressor ZBTB10 and downregulation of microRNA-27a (miR-27a), which constitutively inhibits ZBTB10 expression, and we show for the first time that the effects of betulinic acid on the miR-27a:ZBTB10-Sp transcription factor axis were cannabinoid 1 (CB1) and CB2 receptor-dependent, thus identifying a new cellular target for this anticancer agent.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , MicroRNAs/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Cannabinoid/metabolism , Repressor Proteins/genetics , Triterpenes/pharmacology , YY1 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetinae , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Pentacyclic Triterpenes , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Betulinic Acid
13.
Anticancer Agents Med Chem ; 12(10): 1211-20, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22583404

ABSTRACT

Triterpenoids such as betulinic acid (BA) and synthetic analogs of oleanolic acid [2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO)] and glycyrrhetinic acid [2-cyano-3,11-dioxo-18ß-oleana-1,12-dien-30-oc acid (CDODA)] are potent anticancer agents that exhibit antiproliferative, antiangiogenic, anti-inflammatory and pro-apoptotic activities. Although their effects on multiple pathways have been reported, unifying mechanisms of action have not been reported. Studies in this laboratory have now demonstrated that several triterpenoids including BA and some derivatives, celastrol, methyl ursolate, ß-boswellic acid derivatives, and the synthetic analogs CDDO, CDODA and their esters decreased expression of specificity protein (Sp) transcription factors and several pro-oncogenic Sp-regulated genes in multiple cancer cell lines. The mechanisms of this response are both compound- and cell context-dependent and include activation of both proteasome-dependent and -independent pathways. Triterpenoid-mediated induction of reactive oxygen species (ROS) has now been characterized as an important proteasome-independent pathway for downregulation of Sp transcription factors. ROS decreases expression of microRNA-27a (miR-27a) and miR-20a/miR-17-5p and this results in the induction of the transcriptional "Sp-repressors" ZBTB10 and ZBTB4, respectively, which in turn downregulate Sp and Sp-regulated genes. Triterpenoids also activate or deactive nuclear receptors and G-protein coupled receptors, and these pathways contribute to their antitumorigenic activity and may also play a role in targeting Sp1, Sp3 and Sp4 which are highly overexpressed in multiple cancers and appear to be important for maintaining the cancer phenotype.


Subject(s)
Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Neoplasms/prevention & control , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/pharmacology , Animals , Anticarcinogenic Agents/chemical synthesis , Humans , Molecular Structure , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Pentacyclic Triterpenes/chemical synthesis
14.
Carcinogenesis ; 33(4): 886-94, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22334592

ABSTRACT

Celastrol (CSL) is a naturally occurring triterpenoid acid that exhibits anticancer activity, and in KU7 and 253JB-V bladder cells, CSL induced apoptosis, inhibited growth, colony formation and migration and CSL decreased bladder tumor growth in vivo. CSL also decreased expression of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 and several Sp-regulated genes/proteins including vascular endothelial growth factor, survivin and cyclin D1 and fibroblast growth factor receptor-3, a potential drug target for bladder cancer therapy, has now been characterized as an Sp-regulated gene downregulated by CSL. The mechanism of Sp downregulation by CSL was cell context-dependent due to activation of proteosome-dependent (KU7) and -independent (253JB-V) pathways. In 253JB-V cells, CSL induced reactive oxygen species (ROS) and inhibitors of ROS blocked CSL-induced growth inhibition and repression of Sp1, Sp3 and Sp4. This response was due to induction of the Sp repressors ZBTB10 and ZBTB4 and downregulation of miR-27a and miR-20a/17-5p, respectively, which regulate expression of these transcriptional repressors. Thus, the anticancer activity of CSL in 253JB-V cells is due to induction of ROS and ROS-mediated induction of Sp repressors (ZBTB4/ZBTB10) through downregulation of miR-27a and miR-20a/17-5p.


Subject(s)
Receptor, Fibroblast Growth Factor, Type 3/metabolism , Sp Transcription Factors/metabolism , Triterpenes/pharmacology , Urinary Bladder Neoplasms/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Pentacyclic Triterpenes , Urinary Bladder Neoplasms/pathology
15.
Anticancer Res ; 31(11): 3799-807, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22110202

ABSTRACT

AIM: We hypothesized that the anticancer activity of cannabinoids was linked to induction of phosphatases. MATERIALS AND METHODS: The effects of cannabidiol (CBD) and the synthetic cannabinoid WIN-55,212 (WIN) on LNCaP (prostate) and SW480 (colon) cancer cell proliferation were determined by cell counting; apoptosis was determined by cleavage of poly(ADP)ribose polymerase (PARP) and caspase-3 (Western blots); and phosphatase mRNAs were determined by real-time PCR. The role of phosphatases and cannabinoid receptors in mediating CBD- and WIN-induced apoptosis was determined by inhibition and receptor knockdown. RESULTS: CBD and WIN inhibited LNCaP and SW480 cell growth and induced mRNA expression of several phosphatases, and the phosphatase inhibitor sodium orthovanadate significantly inhibited cannabinoid-induced PARP cleavage in both cell lines, whereas only CBD-induced apoptosis was CB1 and CB2 receptor-dependent. CONCLUSION: Cannabinoid receptor agonists induce phosphatases and phosphatase-dependent apoptosis in cancer cell lines; however, the role of the CB receptor in mediating this response is ligand-dependent.


Subject(s)
Apoptosis/drug effects , Benzoxazines/pharmacology , Cannabidiol/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Morpholines/pharmacology , Naphthalenes/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Apoptosis/physiology , Blotting, Western , Calcium Channel Blockers/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/enzymology , Humans , Male , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/enzymology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism
16.
Nutr Cancer ; 63(7): 1133-42, 2011.
Article in English | MEDLINE | ID: mdl-21919647

ABSTRACT

Ascorbic acid (vitamin C) inhibits cancer cell growth, and there is a controversy regarding the cancer chemoprotective effects of pharmacologic doses of this compound that exhibits prooxidant activity. We hypothesized that the anticancer activity of pharmacologic doses of ascorbic acid (<5 mM) is due, in part, to reactive oxygen species-dependent downregulation of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 and Sp-regulated genes. In this study, ascorbic acid (1-3 mM) decreased RKO and SW480 colon cancer cell proliferation and induced apoptosis and necrosis, and this was accompanied by downregulation of Sp1, Sp3, and Sp4 proteins. In addition, ascorbic acid decreased expression of several Sp-regulated genes that are involved in cancer cell proliferation [hepatocyte growth factor receptor (c-Met), epidermal growth factor receptor and cyclin D1], survival (survivin and bcl-2), and angiogenesis [vascular endothelial growth factor (VEGF) and its receptors (VEGFR1 and VEGFR2)]. Other prooxidants such as hydrogen peroxide exhibited similar activities in colon cancer cells, and cotreatment with glutathione inhibited these responses. This study demonstrates for the first time that the anticancer activities of ascorbic acid are due, in part, to ROS-dependent repression of Sp transcription factors.


Subject(s)
Anticarcinogenic Agents/pharmacology , Ascorbic Acid/pharmacology , Dose-Response Relationship, Drug , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/cytology , Colon/drug effects , Colon/pathology , Colonic Neoplasms/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Reactive Oxygen Species/pharmacology , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/genetics , Sp4 Transcription Factor/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Mol Cancer Res ; 9(2): 195-202, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21156786

ABSTRACT

Ethyl 2-((2,3-bis(nitrooxy)propyl)disulfanyl)benzoate (GT-094) is a novel nitric oxide (NO) chimera containing an nonsteroidal anti-inflammatory drug (NSAID) and NO moieties and also a disulfide pharmacophore that in itself exhibits cancer chemopreventive activity. In this study, the effects and mechanism of action of GT-094 were investigated in RKO and SW480 colon cancer cells. GT-094 inhibited cell proliferation and induced apoptosis in both cell lines and this was accompanied by decreased mitochondrial membrane potential (MMP) and induction of reactive oxygen species (ROS), and these responses were reversed after cotreatment with the antioxidant glutathione. GT-094 also downregulated genes associated with cell growth [cyclin D1, hepatocyte growth factor receptor (c-Met), epidermal growth factor receptor (EGFR)], survival (bcl-2, survivin), and angiogenesis [VEGF and its receptors (VEGFR1 and VEGFR2)]. Results of previous RNA interference studies in this laboratory has shown that these genes are regulated, in part, by specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 that are overexpressed in colon and other cancer cell lines and not surprisingly, GT-094 also decreased Sp1, Sp3, and Sp4 in colon cancer cells. GT-094-mediated repression of Sp and Sp-regulated gene products was due to downregulation of microRNA-27a (miR-27a) and induction of ZBTB10, an Sp repressor that is regulated by miR-27a in colon cancer cells. Moreover, the effects of GT-094 on Sp1, Sp3, Sp4, miR-27a, and ZBTB10 were also inhibited by glutathione suggesting that the anticancer activity of GT-094 in colon cancer cells is due, in part, to activation of an ROS-miR-27a:ZBTB10-Sp transcription factor pathway.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colonic Neoplasms/pathology , DNA-Binding Proteins/metabolism , Disulfides/pharmacology , MicroRNAs/metabolism , Nitrates/pharmacology , Reactive Oxygen Species/metabolism , Repressor Proteins/metabolism , Sp Transcription Factors/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Glutathione/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , MicroRNAs/genetics , Neoplasm Proteins/metabolism , Nitric Oxide/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects
18.
BMC Cancer ; 10: 669, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21129193

ABSTRACT

BACKGROUND: A novel series of methylene-substituted DIMs (C-DIMs), namely 1,1-bis(3'-indolyl)-1-(p-substituted phenyl)methanes containing t-butyl (DIM-C-pPhtBu) and phenyl (DIM-C-pPhC6H5) groups inhibit proliferation of invasive estrogen receptor-negative MDA-MB-231 and MDA-MB-453 human breast cancer cell lines with IC50 values between 1-5 uM. The main purpose of this study was to investigate the pathways of C-DIM-induced cell death. METHODS: The effects of the C-DIMs on apoptotic, necrotic and autophagic cell death were determined using caspase inhibitors, measurement of lactate dehydrogenase release, and several markers of autophagy including Beclin and light chain associated protein 3 expression (LC3). RESULTS: The C-DIM compounds did not induce apoptosis and only DIM-C-pPhCF3 exhibited necrotic effects. However, treatment of MDA-MB-231 and MDA-MB-453 cells with C-DIMs resulted in accumulation of LC3-II compared to LC3-I protein, a characteristic marker of autophagy, and transient transfection of green fluorescent protein-LC3 also revealed that treatment with C-DIMs induced a redistribution of LC3 to autophagosomes after C-DIM treatment. In addition, the autofluorescent drug monodansylcadaverine (MDC), a specific autophagolysosome marker, accumulated in vacuoles after C-DIM treatment, and western blot analysis of lysates from cells treated with C-DIMs showed that the Beclin 1/Bcl-2 protein ratio increased. CONCLUSION: The results suggest that C-DIM compounds may represent a new mechanism-based agent for treating drug-resistant ER-negative breast tumors through induction of autophagy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Indoles/pharmacology , Receptors, Estrogen/analysis , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Blotting, Western , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Female , Humans , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Macrolides/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Necrosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , Xenograft Model Antitumor Assays
19.
J Biol Chem ; 285(33): 25332-44, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20538607

ABSTRACT

Curcumin activates diverse anticancer activities that lead to inhibition of cancer cell and tumor growth, induction of apoptosis, and antiangiogenic responses. In this study, we observed that curcumin inhibits Panc28 and L3.6pL pancreatic cancer cell and tumor growth in nude mice bearing L3.6pL cells as xenografts. In addition, curcumin decreased expression of p50 and p65 proteins and NFkappaB-dependent transactivation and also decreased Sp1, Sp3, and Sp4 transcription factors that are overexpressed in pancreatic cancer cells. Because both Sp transcription factors and NFkappaB regulate several common genes such as cyclin D1, survivin, and vascular endothelial growth factor that contribute to the cancer phenotype, we also investigated interactions between Sp and NFkappaB transcription factors. Results of Sp1, Sp3, and Sp4 knockdown by RNA interference demonstrate that both p50 and p65 are Sp-regulated genes and that inhibition of constitutive or tumor necrosis factor-induced NFkappaB by curcumin is dependent on down-regulation of Sp1, Sp3, and Sp4 proteins by this compound. Curcumin also decreased mitochondrial membrane potential and induced reactive oxygen species in pancreatic cancer cells, and this pathway is required for down-regulation of Sp proteins in these cells, demonstrating that the mitochondriotoxic effects of curcumin are important for its anticancer activities.


Subject(s)
Curcumin/therapeutic use , NF-kappa B/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Sp Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/pharmacology , Cyclin D1/genetics , Electrophoretic Mobility Shift Assay , Electrophysiology , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , NF-kappa B/genetics , Polymerase Chain Reaction , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Sp Transcription Factors/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/genetics , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Xenograft Model Antitumor Assays
20.
Exp Cell Res ; 316(13): 2174-88, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20435036

ABSTRACT

Arsenic trioxide exhibits antiproliferative, antiangiogenic and proapoptotic activity in cancer cells, and many genes associated with these responses are regulated by specificity protein (Sp) transcription factors. Treatment of cancer cells derived from urologic (bladder and prostate) and gastrointestinal (pancreas and colon) tumors with arsenic trioxide demonstrated that these cells exhibited differential responsiveness to the antiproliferative effects of this agent and this paralleled their differential repression of Sp1, Sp3 and Sp4 proteins in the same cell lines. Using arsenic trioxide-responsive KU7 and non-responsive 253JB-V bladder cancer cells as models, we show that in KU7 cells, < or =5 microM arsenic trioxide decreased Sp1, Sp3 and Sp4 and several Sp-dependent genes and responses including cyclin D1, epidermal growth factor receptor, bcl-2, survivin and vascular endothelial growth factor, whereas at concentrations up to 15 microM, minimal effects were observed in 253JB-V cells. Arsenic trioxide also inhibited tumor growth in athymic mice bearing KU7 cells as xenografts, and expression of Sp1, Sp3 and Sp4 was significantly decreased. Inhibitors of oxidative stress such as glutathione or dithiothreitol protected KU7 cells from arsenic trioxide-induced antiproliferative activity and Sp repression, whereas glutathione depletion sensitized 253JB-V cells to arsenic trioxide. Mechanistic studies suggested that arsenic trioxide-dependent downregulation of Sp and Sp-dependent genes was due to decreased mitochondrial membrane potential and induction of reactive oxygen species, and the role of peroxides in mediating these responses was confirmed using hydrogen peroxide.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Oxides/pharmacology , Sp Transcription Factors/genetics , Urinary Bladder Neoplasms/pathology , Animals , Apoptosis/drug effects , Arsenic Trioxide , Blotting, Western , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Humans , Mice , Mice, Nude , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sp Transcription Factors/metabolism , Tumor Cells, Cultured , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...