39 results on '"Jutooru I"'
Search Results
2. HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer
- Author
-
Kim, K, Jutooru, I, Chadalapaka, G, Johnson, G, Frank, J, Burghardt, R, Kim, S, and Safe, S
- Published
- 2013
- Full Text
- View/download PDF
3. Estrogenic Endocrine Disruptors: Molecular Characteristics and Human Impacts
- Author
-
Safe, S., primary, Jutooru, I., additional, and Chadalapaka, G., additional
- Published
- 2010
- Full Text
- View/download PDF
4. In vitro evaluation of the non-steroidal anti-inflammatory drug, tolfenamic acid, for canine solid tumours
- Author
-
Wilson-Robles, H., Chadalapaka, G., Jutooru, I., Sheppard, S., and Safe, S.
- Published
- 2011
5. Critical analysis of the potential for the therapeutic targeting of the Sp1 transcription factor in pancreatic cancer
- Author
-
Jutooru I, Chadalapaka G, and Safe S
- Subjects
lcsh:Diseases of the digestive system. Gastroenterology ,lcsh:RC799-869 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 - Abstract
Indira Jutooru,1 Gayathri Chadalapaka,1 Stephen Safe1,21Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA; 2Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USAAbstract: Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer-related deaths in developed countries and, in 2013, it is estimated that in excess of 45,220 new cases were diagnosed in the United States. PDAC is a highly aggressive disease that invariably evades early diagnosis. The mean survival time for patients with metastatic disease is only 3–6 months, and only 20%–30% of pancreatic cancer patients are alive after 12 months. Because pancreatic cancers are frequently detected at an advanced stage, treatments have provided very limited improvements in tumor regression and overall survival times after diagnosis. 5-Fluorouracil alone or in combination with other drugs has been extensively used for treatment of advanced pancreatic cancer, and gemcitabine has partially replaced 5-fluorouracil as a treatment for pancreatic cancer. Gemcitabine provides increased clinical benefits in terms of response rate; however, future studies need to focus on developing treatment modalities that will improve the survival rate for pancreatic cancer patients. Specificity protein 1 (Sp1) is overexpressed in PDAC patients, and high expression is associated with poor prognosis, lymph node metastasis, and low survival. Knockdown studies have shown that Sp1 plays an important role in cell growth, angiogenesis, inflammation, survival, and metastasis. Sp1 expression is low in normal tissue when compared to tumor tissue, which makes Sp1 a potential target for development of new mechanism-based drugs for treatment of pancreatic cancer. Several drugs such as tolfenamic acid, betulinic acid, and methyl-2-cyano3,12-dioxooleana-1,9(11)-dien-28-oate are shown to downregulate Sp1 expression through various pathways. This review summarizes the role of Sp1 in pancreatic cancer and delineates the mechanisms of action of various drugs that downregulate expression of Sp1 and other Sp transcription factors.Keywords: Sp transcription factors, microRNAs, ZBTB repressors, ROS
- Published
- 2014
6. Estrogenic Endocrine Disruptors: Molecular Characteristics and Human Impacts
- Author
-
Safe, S., Jutooru, I., Jin, U.-H., and Chadalapaka, G.
- Published
- 2015
- Full Text
- View/download PDF
7. Metformin inhibits pancreatic cancer cell and tumor growth and downregulates Sp transcription factors
- Author
-
Nair, V., primary, Pathi, S., additional, Jutooru, I., additional, Sreevalsan, S., additional, Basha, R., additional, Abdelrahim, M., additional, Samudio, I., additional, and Safe, S., additional
- Published
- 2013
- Full Text
- View/download PDF
8. HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer
- Author
-
Kim, K, primary, Jutooru, I, additional, Chadalapaka, G, additional, Johnson, G, additional, Frank, J, additional, Burghardt, R, additional, Kim, S, additional, and Safe, S, additional
- Published
- 2012
- Full Text
- View/download PDF
9. Effect of Tolfenamic Acid on Canine Cancer Cell Proliferation, Specificity Protein (Sp) Transcription Factors, and Sp-Regulated Proteins in Canine Osteosarcoma, Mammary Carcinoma, and Melanoma Cells
- Author
-
Wilson, H., primary, Chadalapaka, G., additional, Jutooru, I., additional, Sheppard, S., additional, Pfent, C., additional, and Safe, S., additional
- Published
- 2012
- Full Text
- View/download PDF
10. 2-Cyano-lup-1-en-3-oxo-20-oic acid, a cyano derivative of betulinic acid, activates peroxisome proliferator-activated receptor in colon and pancreatic cancer cells
- Author
-
Chintharlapalli, S., primary, Papineni, S., additional, Liu, S., additional, Jutooru, I., additional, Chadalapaka, G., additional, Cho, S.-d., additional, Murthy, R. S., additional, You, Y., additional, and Safe, S., additional
- Published
- 2007
- Full Text
- View/download PDF
11. 1,1-Bis(3'-indolyl)-1-(p-substituted phenyl)methanes induce autophagic cell death in estrogen receptor negative breast cancer
- Author
-
Chadalapaka Gayathri, Barhoumi Rola, Burghardt Robert C, Frankel Arthur E, Su Yunpeng, Vanderlaag Kathy, Jutooru Indira, and Safe Stephen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
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.
- Published
- 2010
- Full Text
- View/download PDF
12. Reactive Oxygen Species (ROS)-Inducing Triterpenoid Inhibits Rhabdomyosarcoma Cell and Tumor Growth through Targeting Sp Transcription Factors.
- Author
-
Kasiappan R, Jutooru I, Mohankumar K, Karki K, Lacey A, and Safe S
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Female, Humans, Mice, Mice, Nude, Rhabdomyosarcoma pathology, Transfection, Triterpenes pharmacology, Reactive Oxygen Species metabolism, Rhabdomyosarcoma drug therapy, Sp Transcription Factors genetics, Triterpenes therapeutic use
- Abstract
Methyl 2-trifluoromethyl-3,11-dioxo-18β-olean-1,12-dien-3-oate (CF
3 DODA-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. CF3 DODA-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. CF3 DODA-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 CF3 DODA-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 CF3 DODA-Me further emphasize the sensitivity of RMS cells to ROS inducers and their potential clinical applications for treating this deadly disease. IMPLICATIONS: CF3 DODA-Me and HDAC inhibitors that induce ROS-dependent Sp downregulation could be developed for clinical applications in treating rhabdomyosarcoma., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
13. Role of metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1) in pancreatic cancer.
- Author
-
Cheng Y, Imanirad P, Jutooru I, Hedrick E, Jin UH, Rodrigues Hoffman A, Leal de Araujo J, Morpurgo B, Golovko A, and Safe S
- Subjects
- Animals, Cell Line, Tumor, Down-Regulation, Gene Expression Regulation, Neoplastic, Gene Knockdown Techniques, Humans, Mice, Mice, Transgenic, Pancreatic Neoplasms genetics, Biomarkers, Tumor genetics, Pancreatic Neoplasms pathology, RNA, Long Noncoding genetics
- 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.
- Published
- 2018
- Full Text
- View/download PDF
14. Benzyl Isothiocyanate (BITC) Induces Reactive Oxygen Species-dependent Repression of STAT3 Protein by Down-regulation of Specificity Proteins in Pancreatic Cancer.
- Author
-
Kasiappan R, Jutooru I, Karki K, Hedrick E, and Safe S
- Subjects
- 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, 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
- 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., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
15. The long non-coding RNA HOTTIP enhances pancreatic cancer cell proliferation, survival and migration.
- Author
-
Cheng Y, Jutooru I, Chadalapaka G, Corton JC, and Safe S
- Subjects
- 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, Cell Movement, Cell Proliferation, Pancreatic Neoplasms metabolism, RNA, Long Noncoding metabolism
- 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.
- Published
- 2015
- Full Text
- View/download PDF
16. The transcriptional repressor ZBTB4 regulates EZH2 through a MicroRNA-ZBTB4-specificity protein signaling axis.
- Author
-
Yang WS, Chadalapaka G, Cho SG, Lee SO, Jin UH, Jutooru I, Choi K, Leung YK, Ho SM, Safe S, and Kim K
- Subjects
- 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, Breast Neoplasms genetics, Gene Expression Regulation, Neoplastic physiology, MicroRNAs genetics, Polycomb Repressive Complex 2 genetics, Repressor Proteins genetics
- 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., (Published by Elsevier Inc.)
- Published
- 2014
- Full Text
- View/download PDF
17. Mechanism of action of phenethylisothiocyanate and other reactive oxygen species-inducing anticancer agents.
- Author
-
Jutooru I, Guthrie AS, Chadalapaka G, Pathi S, Kim K, Burghardt R, Jin UH, and Safe S
- Subjects
- 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, Anticarcinogenic Agents pharmacology, Enzyme Inhibitors pharmacology, Isothiocyanates pharmacology, Reactive Oxygen Species metabolism
- 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., (Copyright © 2014, American Society for Microbiology. All Rights Reserved.)
- Published
- 2014
- Full Text
- View/download PDF
18. Transcription factor Sp1, also known as specificity protein 1 as a therapeutic target.
- Author
-
Safe S, Imanirad P, Sreevalsan S, Nair V, and Jutooru I
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Down-Regulation, Humans, Neoplasms drug therapy, Neoplasms metabolism, Sp1 Transcription Factor metabolism
- 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.
- Published
- 2014
- Full Text
- View/download PDF
19. The drug resistance suppression induced by curcuminoids in colon cancer SW-480 cells is mediated by reactive oxygen species-induced disruption of the microRNA-27a-ZBTB10-Sp axis.
- Author
-
Noratto GD, Jutooru I, Safe S, Angel-Morales G, and Mertens-Talcott SU
- Subjects
- 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, Colonic Neoplasms metabolism, Curcumin pharmacology, Gene Expression Regulation, Neoplastic, MicroRNAs genetics, Reactive Oxygen Species metabolism
- 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., (© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2013
- Full Text
- View/download PDF
20. Inhibition of rhabdomyosarcoma cell and tumor growth by targeting specificity protein (Sp) transcription factors.
- Author
-
Chadalapaka G, Jutooru I, Sreevalsan S, Pathi S, Kim K, Chen C, Crose L, Linardic C, and Safe S
- Subjects
- 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, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Gene Expression Regulation, Neoplastic drug effects, Rhabdomyosarcoma drug therapy, Rhabdomyosarcoma metabolism, Sp Transcription Factors metabolism, ortho-Aminobenzoates pharmacology
- 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., (Copyright © 2012 UICC.)
- Published
- 2013
- Full Text
- View/download PDF
21. Unifying mechanisms of action of the anticancer activities of triterpenoids and synthetic analogs.
- Author
-
Safe SH, Prather PL, Brents LK, Chadalapaka G, and Jutooru I
- Subjects
- Animals, Anticarcinogenic Agents chemical synthesis, Humans, Molecular Structure, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Pentacyclic Triterpenes chemical synthesis, Anticarcinogenic Agents chemistry, Anticarcinogenic Agents pharmacology, Neoplasms prevention & control, Pentacyclic Triterpenes chemistry, Pentacyclic Triterpenes pharmacology
- 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-18β-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, β-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.
- Published
- 2012
- Full Text
- View/download PDF
22. Betulinic acid targets YY1 and ErbB2 through cannabinoid receptor-dependent disruption of microRNA-27a:ZBTB10 in breast cancer.
- Author
-
Liu X, Jutooru I, Lei P, Kim K, Lee SO, Brents LK, Prather PL, and Safe S
- Subjects
- 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, 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
- 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., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
23. Celastrol decreases specificity proteins (Sp) and fibroblast growth factor receptor-3 (FGFR3) in bladder cancer cells.
- Author
-
Chadalapaka G, Jutooru I, and Safe S
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Mice, Mice, Nude, Pentacyclic Triterpenes, Urinary Bladder Neoplasms pathology, Receptor, Fibroblast Growth Factor, Type 3 metabolism, Sp Transcription Factors metabolism, Triterpenes pharmacology, Urinary Bladder Neoplasms metabolism
- 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.
- Published
- 2012
- Full Text
- View/download PDF
24. Aspirin inhibits colon cancer cell and tumor growth and downregulates specificity protein (Sp) transcription factors.
- Author
-
Pathi S, Jutooru I, Chadalapaka G, Nair V, Lee SO, and Safe S
- Subjects
- 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, Aspirin therapeutic use, Colonic Neoplasms drug therapy, Colonic Neoplasms metabolism, Sp Transcription Factors metabolism
- 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 β-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.
- Published
- 2012
- Full Text
- View/download PDF
25. Induction of apoptosis by cannabinoids in prostate and colon cancer cells is phosphatase dependent.
- Author
-
Sreevalsan S, Joseph S, Jutooru I, Chadalapaka G, and Safe SH
- Subjects
- 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, 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
- 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.
- Published
- 2011
26. GT-094, a NO-NSAID, inhibits colon cancer cell growth by activation of a reactive oxygen species-microRNA-27a: ZBTB10-specificity protein pathway.
- Author
-
Pathi SS, Jutooru I, Chadalapaka G, Sreevalsan S, Anand S, Thatcher GR, and Safe S
- Subjects
- 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, 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
- 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.
- Published
- 2011
- Full Text
- View/download PDF
27. Pharmacologic doses of ascorbic acid repress specificity protein (Sp) transcription factors and Sp-regulated genes in colon cancer cells.
- Author
-
Pathi SS, Lei P, Sreevalsan S, Chadalapaka G, Jutooru I, and Safe S
- Subjects
- 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, Anticarcinogenic Agents pharmacology, Ascorbic Acid pharmacology, Dose-Response Relationship, Drug
- 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.
- Published
- 2011
- Full Text
- View/download PDF
28. 1,1-Bis(3'-indolyl)-1-(p-substituted phenyl)methanes induce autophagic cell death in estrogen receptor negative breast cancer.
- Author
-
Vanderlaag K, Su Y, Frankel AE, Burghardt RC, Barhoumi R, Chadalapaka G, Jutooru I, and Safe S
- Subjects
- 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, Antineoplastic Agents pharmacology, Autophagy drug effects, Biomarkers, Tumor analysis, Breast Neoplasms drug therapy, Indoles pharmacology, Receptors, Estrogen analysis
- 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.
- Published
- 2010
- Full Text
- View/download PDF
29. Inhibition of NFkappaB and pancreatic cancer cell and tumor growth by curcumin is dependent on specificity protein down-regulation.
- Author
-
Jutooru I, Chadalapaka G, Lei P, and Safe S
- Subjects
- 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, Curcumin therapeutic use, NF-kappa B metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, Sp Transcription Factors metabolism
- 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.
- Published
- 2010
- Full Text
- View/download PDF
30. Methyl 2-cyano-3,12-dioxooleana-1,9-dien-28-oate decreases specificity protein transcription factors and inhibits pancreatic tumor growth: role of microRNA-27a.
- Author
-
Jutooru I, Chadalapaka G, Abdelrahim M, Basha MR, Samudio I, Konopleva M, Andreeff M, and Safe S
- Subjects
- Animals, Base Sequence, Blotting, Western, Cell Line, Tumor, Cell Proliferation, DNA Primers, Humans, Male, Mice, Mice, Nude, Oleanolic Acid pharmacology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Polymerase Chain Reaction, Reactive Oxygen Species metabolism, MicroRNAs metabolism, Oleanolic Acid analogs & derivatives, Pancreatic Neoplasms pathology, Transcription Factors metabolism
- Abstract
The anticancer agent 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) and its methyl ester (CDDO-Me) typically induce a broad spectrum of growth-inhibitory, proapoptotic, and antiangiogenic responses. Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). RNA interference studies indicate that these repressed genes are regulated by specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and Western blot analysis of lysates from pancreatic cancer cells treated with CDDO and CDDO-Me shows for the first time that both compounds decreased the expression of Sp1, Sp3, and Sp4. Moreover, CDDO-Me (7.5 mg/kg/day) also inhibited pancreatic human L3.6pL tumor growth and down-regulated Sp1, Sp3, and Sp4 in tumors using an orthotopic pancreatic cancer model. CDDO-Me also induced reactive oxygen species (ROS) and decreased mitochondrial membrane potential (MMP) in Panc1 and L3.6pL cells, and cotreatment with antioxidants (glutathione and dithiothreitol) blocked the formation of ROS, reversed the loss of MMP, and inhibited down-regulation of Sp1, Sp3, and Sp4. Repression of Sp and Sp-dependent genes by CDDO-Me was due to the down-regulation of microRNA-27a and induction of zinc finger and BTB domain containing 10 (ZBTB10), an Sp repressor, and these responses were also reversed by antioxidants. Thus, the anticancer activity of CDDO-Me is due, in part, to activation of ROS, which in turn targets the microRNA-27a:ZBTB10-Sp transcription factor axis. This results in decreased expression of Sp-regulated genes, growth inhibition, induction of apoptosis, and antiangiogenic responses.
- Published
- 2010
- Full Text
- View/download PDF
31. Arsenic trioxide downregulates specificity protein (Sp) transcription factors and inhibits bladder cancer cell and tumor growth.
- Author
-
Jutooru I, Chadalapaka G, Sreevalsan S, Lei P, Barhoumi R, Burghardt R, and Safe S
- Subjects
- 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, Antineoplastic Agents pharmacology, Arsenicals pharmacology, Gene Expression Regulation, Neoplastic drug effects, Oxides pharmacology, Sp Transcription Factors genetics, Urinary Bladder Neoplasms pathology
- 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., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
32. Drugs that target specificity proteins downregulate epidermal growth factor receptor in bladder cancer cells.
- Author
-
Chadalapaka G, Jutooru I, Burghardt R, and Safe S
- Subjects
- Autophagy drug effects, Autophagy physiology, Curcumin pharmacology, Down-Regulation physiology, Drug Delivery Systems methods, ErbB Receptors genetics, ErbB Receptors metabolism, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Neoplastic physiology, Humans, Microtubule-Associated Proteins metabolism, Pentacyclic Triterpenes, Proto-Oncogene Proteins c-akt metabolism, RNA Stability, RNA, Small Interfering pharmacology, Sp Transcription Factors genetics, Sp1 Transcription Factor genetics, Sp1 Transcription Factor metabolism, Sp3 Transcription Factor metabolism, Triterpenes pharmacology, Urinary Bladder Neoplasms pathology, Betulinic Acid, Antineoplastic Agents pharmacology, Down-Regulation drug effects, ErbB Receptors antagonists & inhibitors, Sp Transcription Factors metabolism, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms metabolism
- Abstract
The epidermal growth factor receptor (EGFR) is an important chemotherapeutic target for tyrosine kinase inhibitors and antibodies that block the extracellular domain of EGFR. Betulinic acid (BA) and curcumin inhibited bladder cancer cell growth and downregulated specificity protein (Sp) transcription factors, and this was accompanied by decreased expression of EGFR mRNA and protein levels. EGFR, a putative Sp-regulated gene, was also decreased in cells transfected with a cocktail (iSp) containing small inhibitory RNAs for Sp1, Sp3, and Sp4, and RNA interference with individual Sp knockdown indicated that EGFR expression was primarily regulated by Sp1 and Sp3. BA, curcumin, and iSp also decreased phosphorylation of Akt in these cells, and downregulation of EGFR by BA, curcumin, and iSp was accompanied by induction of LC3 and autophagy, which is consistent with recent studies showing that EGFR suppresses autophagic cell death. The results show that EGFR is an Sp-regulated gene in bladder cancer, and drugs such as BA and curcumin that repress Sp proteins also ablate EGFR expression. Thus, compounds such as curcumin and BA that downregulate Sp transcription factors represent a novel class of anticancer drugs that target EGFR in bladder cancer cells and tumors by inhibiting receptor expression., ((c)2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
33. 1,1-Bis(3'-indolyl)-1-(p-bromophenyl)methane and related compounds repress survivin and decrease gamma-radiation-induced survivin in colon and pancreatic cancer cells.
- Author
-
Sreevalsan S, Jutooru I, Chadalapaka G, Walker M, and Safe S
- Subjects
- Apoptosis drug effects, Apoptosis radiation effects, Blotting, Western, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation radiation effects, Down-Regulation, Gamma Rays, Gene Expression drug effects, Gene Expression radiation effects, Humans, Inhibitor of Apoptosis Proteins, Microtubule-Associated Proteins biosynthesis, Pancreatic Neoplasms metabolism, RNA, Messenger analysis, Reverse Transcriptase Polymerase Chain Reaction, Survivin, Transfection, Antineoplastic Agents pharmacology, Indoles pharmacology, Microtubule-Associated Proteins drug effects, Microtubule-Associated Proteins radiation effects, Pancreatic Neoplasms genetics, Radiation Tolerance genetics
- Abstract
1,1-Bis(3'-indolyl)-1-(p-bromophenyl)methane (DIM-C-pPhBr) and the 2,2'-dimethyl analog (2,2'-diMeDIM-C-pPhBr) inhibit proliferation and induce apoptosis in SW480 colon and Panc28 pancreatic cancer cells. In this study, treatment with 10-20 microM concentrations of these compounds for 24 h induced cleaved PARP and decreased survivin protein and mRNA expression in both cell lines. However, results of time course studies show that DIM-C-pPhBr and 2,2'-diMeDIM-C-pPhBr decrease survivin protein within 2 h after treatment, whereas survivin mRNA levels were decreased only at later time-points indicating activation of transcription-independent and -dependent pathways for downregulation of survivin. In addition, we also observed that gamma-radiation inhibited pancreatic and colon cancer cell growth and this was associated with enhanced expression of survivin after 24 (SW480) or 24 and 48 h (Panc28) and correlated with previous studies on the role of survivin in radiation-resistance. However, in cells co-treated with gamma-radiation plus DIM-C-pPhBr or 2,2'-diMeDIM-C-pPhBr, induction of survivin by gamma-radiation was inhibited after co-treatment with both compounds, suggesting applications for these drugs in combination cancer chemotherapy with gamma-radiation.
- Published
- 2009
- Full Text
- View/download PDF
34. Oncogenic microRNA-27a is a target for anticancer agent methyl 2-cyano-3,11-dioxo-18beta-olean-1,12-dien-30-oate in colon cancer cells.
- Author
-
Chintharlapalli S, Papineni S, Abdelrahim M, Abudayyeh A, Jutooru I, Chadalapaka G, Wu F, Mertens-Talcott S, Vanderlaag K, Cho SD, Smith R 3rd, and Safe S
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Blotting, Northern, Blotting, Western, Cell Cycle, Cell Proliferation drug effects, Colonic Neoplasms pathology, Glycyrrhetinic Acid chemical synthesis, Glycyrrhetinic Acid chemistry, Glycyrrhetinic Acid pharmacology, Humans, Mice, Mice, Nude, MicroRNAs genetics, PPAR gamma agonists, PPAR gamma metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering pharmacology, Reverse Transcriptase Polymerase Chain Reaction, Sp Transcription Factors antagonists & inhibitors, Sp Transcription Factors genetics, Sp Transcription Factors metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Colonic Neoplasms drug therapy, Colonic Neoplasms genetics, Gene Expression Regulation, Neoplastic drug effects, Glycyrrhetinic Acid analogs & derivatives, MicroRNAs metabolism, Oncogenes
- Abstract
Methyl 2-cyano-3,11-dioxo-18beta-olean-1,12-dien-30-oate (CDODA-Me) is a synthetic derivative of glycyrrhetinic acid, a triterpenoid phytochemical found in licorice extracts. CDODA-Me inhibited growth of RKO and SW480 colon cancer cells and this was accompanied by decreased expression of Sp1, Sp3 and Sp4 protein and mRNA and several Sp-dependent genes including survivin, vascular endothelial growth factor (VEGF), and VEGF receptor 1 (VEGFR1 or Flt-1). CDODA-Me also induced apoptosis, arrested RKO and SW480 cells at G(2)/M, and inhibited tumor growth in athymic nude mice bearing RKO cells as xenografts. CDODA-Me decreased expression of microRNA-27a (miR-27a), and this was accompanied by increased expression of 2 miR-27a-regulated mRNAs, namely ZBTB10 (an Sp repressor) and Myt-1 which catalyzes phosphorylation of cdc2 to inhibit progression of cells through G(2)/M. Both CDODA-Me and antisense miR-27a induced comparable responses in RKO and SW480 cells, suggesting that the potent anticarcinogenic activity of CDODA-Me is due to repression of oncogenic miR-27a.
- Published
- 2009
- Full Text
- View/download PDF
35. Induction of apoptosis and nonsteroidal anti-inflammatory drug-activated gene 1 in pancreatic cancer cells by a glycyrrhetinic acid derivative.
- Author
-
Jutooru I, Chadalapaka G, Chintharlapalli S, Papineni S, and Safe S
- Subjects
- Activating Transcription Factor 3 metabolism, Adipose Tissue cytology, Adipose Tissue drug effects, Adipose Tissue metabolism, Animals, Blotting, Western, Cell Cycle Proteins metabolism, Cell Differentiation drug effects, Cell Proliferation drug effects, Early Growth Response Protein 1 metabolism, Glycyrrhetinic Acid analogs & derivatives, Humans, Luciferases metabolism, Mice, Mitogen-Activated Protein Kinase 1 metabolism, PPAR gamma metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms metabolism, Phosphatidylinositol 3-Kinases metabolism, Promoter Regions, Genetic, Signal Transduction, Transfection, Tumor Cells, Cultured, p38 Mitogen-Activated Protein Kinases metabolism, Apoptosis drug effects, Glycyrrhetinic Acid pharmacology, Growth Differentiation Factor 15 metabolism, Pancreatic Neoplasms pathology
- Abstract
Methyl 2-cyano-3,11-dioxo-18beta-olean-1,12-dien-30-oate (CDODA-Me) is a synthetic triterpenoid derived from glycyrrhetinic acid, a bioactive phytochemical in licorice, CDODA-Me inhibits growth of Panc1 and Panc28 pancreatic cancer cell lines and activates peroxisome proliferator-activated receptor gamma (PPARgamma)-dependent transactivation in these cells. CDODA-Me also induced p21 and p27 protein expression and downregulates cyclin D1; however, these responses were receptor-independent. CDODA-Me induced apoptosis in Panc1 and Panc28 cells, and this was accompanied by receptor-independent induction of the proapoptotic proteins early growth response-1 (Egr-1), nonsteroidal anti-inflammatory drug-activated gene-1 (NAG-1), and activating transcription factor-3 (ATF3). Induction of NAG-1 and Egr-1 by CDODA-Me was dependent on activation of phosphatidylinositol-3-kinase (PI3-K) and/or p42 and p38 mitogen-activated protein kinase (MAPK) pathways but there were differences between Panc28 and Panc1 cells. Induction of NAG-1 in Panc28 cells was p38-MAPK- and PI3-K-dependent but Egr-1-independent, whereas induction in Panc1 cells was associated with activation of p38-MAPK, PI3-K, and p42-MAPK and was only partially Egr-1-dependent. This is the first report of the induction of the proapoptotic protein NAG-1 in pancreatic cancer cells.
- Published
- 2009
- Full Text
- View/download PDF
36. Curcumin decreases specificity protein expression in bladder cancer cells.
- Author
-
Chadalapaka G, Jutooru I, Chintharlapalli S, Papineni S, Smith R 3rd, Li X, and Safe S
- Subjects
- Animals, Antineoplastic Agents, Phytogenic pharmacology, Antineoplastic Agents, Phytogenic therapeutic use, Cell Cycle drug effects, Cell Cycle genetics, Dose-Response Relationship, Drug, Down-Regulation drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Mice, Mice, Nude, RNA, Small Interfering pharmacology, Sp Transcription Factors antagonists & inhibitors, Time Factors, Tumor Burden drug effects, Tumor Cells, Cultured, Urinary Bladder Neoplasms pathology, Xenograft Model Antitumor Assays, Curcumin pharmacology, Curcumin therapeutic use, Sp Transcription Factors genetics, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms genetics
- Abstract
Curcumin is the active component of tumeric, and this polyphenolic compound has been extensively investigated as an anticancer drug that modulates multiple pathways and genes. In this study, 10 to 25 micromol/L curcumin inhibited 253JB-V and KU7 bladder cancer cell growth, and this was accompanied by induction of apoptosis and decreased expression of the proapoptotic protein survivin and the angiogenic proteins vascular endothelial growth factor (VEGF) and VEGF receptor 1 (VEGFR1). Because expression of survivin, VEGF, and VEGFR1 are dependent on specificity protein (Sp) transcription factors, we also investigated the effects of curcumin on Sp protein expression as an underlying mechanism for the apoptotic and antiangiogenic activity of this compound. The results show that curcumin induced proteasome-dependent down-regulation of Sp1, Sp3, and Sp4 in 253JB-V and KU7 cells. Moreover, using RNA interference with small inhibitory RNAs for Sp1, Sp3, and Sp4, we observed that curcumin-dependent inhibition of nuclear factor kappaB (NF-kappaB)-dependent genes, such as bcl-2, survivin, and cyclin D1, was also due, in part, to loss of Sp proteins. Curcumin also decreased bladder tumor growth in athymic nude mice bearing KU7 cells as xenografts and this was accompanied by decreased Sp1, Sp3, and Sp4 protein levels in tumors. These results show for the first time that one of the underlying mechanisms of action of curcumin as a cancer chemotherapeutic agent is due, in part, to decreased expression of Sp transcription factors in bladder cancer cells.
- Published
- 2008
- Full Text
- View/download PDF
37. Structure-dependent inhibition of bladder and pancreatic cancer cell growth by 2-substituted glycyrrhetinic and ursolic acid derivatives.
- Author
-
Chadalapaka G, Jutooru I, McAlees A, Stefanac T, and Safe S
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Glycyrrhetinic Acid chemistry, Humans, Methylation, Molecular Structure, Structure-Activity Relationship, Triterpenes chemistry, Ursolic Acid, Glycyrrhetinic Acid chemical synthesis, Glycyrrhetinic Acid pharmacology, Pancreatic Neoplasms pathology, Triterpenes chemical synthesis, Triterpenes pharmacology, Urinary Bladder Neoplasms pathology
- Abstract
Derivatives of oleanolic acid, ursolic acid and glycyrrhetinic acid substituted with electron-withdrawing groups at the 2-position in the A-ring which also contains a 1-en-3-one structure are potent inhibitors of cancer cell growth. In this study, we have compared the effects of several 2-substituted analogs of triterpenoid acid methyl esters derived from ursolic and glycyrrhetinic acid on proliferation of KU7 and 253JB-V bladder and Panc-1 and Panc-28 pancreatic cancer cells. The results show that the 2-cyano and 2-trifluoromethyl derivatives were the most active compounds. The glycyrrhetinic acid derivatives with the rearranged C-ring containing the 9(11)-en-12-one structure were generally more active than the corresponding 12-en-11-one isomers. However, differences in growth inhibitory IC(50) values were highly variable and dependent on the 2-substituent (CN vs CF(3)) and cancer cell context.
- Published
- 2008
- Full Text
- View/download PDF
38. 2-cyano-lup-1-en-3-oxo-20-oic acid, a cyano derivative of betulinic acid, activates peroxisome proliferator-activated receptor gamma in colon and pancreatic cancer cells.
- Author
-
Chintharlapalli S, Papineni S, Liu S, Jutooru I, Chadalapaka G, Cho SD, Murthy RS, You Y, and Safe S
- Subjects
- 3T3-L1 Cells, Animals, Cell Differentiation drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Colonic Neoplasms pathology, Drug Screening Assays, Antitumor, Humans, Kruppel-Like Factor 4, Mice, Pancreatic Neoplasms pathology, Colonic Neoplasms metabolism, PPAR gamma agonists, Pancreatic Neoplasms metabolism, Triterpenes pharmacology
- Abstract
Betulinic acid (BA) is a phytochemical triterpenoid acid from bark extracts and is cytotoxic to cancer cells and tumors. We modified the A-ring of BA to give a 2-cyano-1-en-3-one moiety and the effects of the 2-cyano-lup-1-en-3-oxo-20-oic acid (CN-BA), 2-cyano derivative of BA, and its methyl ester (CN-BA-Me) were investigated in colon and pancreatic cancer cells. Both CN-BA and CN-BA-Me were highly cytotoxic to Panc-28 pancreatic and SW480 colon cancer cells. CN-BA and CN-BA-Me also induced differentiation in 3T3-L1 adipocytes, which exhibited a characteristic fat droplet accumulation induced by peroxisome proliferator-activated receptor gamma (PPARgamma) agonists. Based on these results, we investigated the activities of CN-BA and CN-BA-Me as PPARgamma agonists using several receptor-mediated responses including activation of transfected PPARgamma-responsive constructs, induction of p21 in Panc-28 cells and induction of caveolin-1 and Krüppel-like factor 4 in colon cancer cells. The results clearly demonstrated that both CN-BA and CN-BA-Me activated PPARgamma-dependent responses in colon (caveolin-1) and pancreatic (p21) cancer cells, whereas induction of KLF4 by these compounds in colon cancer cells was PPARgamma independent and also dependent on cell context. The PPARgamma agonist activities of CN-BA and CN-BA-Me were structure-, response/gene- and cell context-dependent suggesting that these compounds are a novel class of selective PPARgamma modulators with potential for clinical treatment of colon and pancreatic cancer.
- Published
- 2007
- Full Text
- View/download PDF
39. Structure-dependent activity of glycyrrhetinic acid derivatives as peroxisome proliferator-activated receptor {gamma} agonists in colon cancer cells.
- Author
-
Chintharlapalli S, Papineni S, Jutooru I, McAlees A, and Safe S
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis, Cell Line, Tumor, Cell Proliferation, Glycyrrhiza, Humans, Kruppel-Like Factor 4, Ligands, Models, Chemical, PPAR gamma antagonists & inhibitors, Plasmids metabolism, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Two-Hybrid System Techniques, Colonic Neoplasms drug therapy, Glycyrrhetinic Acid analogs & derivatives, Glycyrrhetinic Acid chemistry, PPAR gamma agonists
- Abstract
Glycyrrhizin, a pentacyclic triterpene glycoside, is the major phytochemical in licorice. This compound and its hydrolysis product glycyrrhetinic acid have been associated with the multiple therapeutic properties of licorice extracts. We have investigated the effects of 2-cyano substituted analogues of glycyrrhetinic acid on their cytotoxicities and activity as selective peroxisome proliferator-activated receptor gamma (PPARgamma) agonists. Methyl 2-cyano-3,11-dioxo-18beta-olean-1,12-dien-30-oate (beta-CDODA-Me) and methyl 2-cyano-3,11-dioxo-18alpha-olean-1,12-dien-30-oate (alpha-CDODA-Me) were more cytotoxic to colon cancer cells than their des-cyano analogues and introduction of the 2-cyano group into the pentacyclic ring system was necessary for the PPARgamma agonist activity of alpha-CDODA-Me and beta-CDODA-Me isomers. However, in mammalian two-hybrid assays, both compounds differentially induced interactions of PPARgamma with coactivators, suggesting that these isomers, which differ only in the stereochemistry at C18 which affects conformation of the E-ring, are selective receptor modulators. This selectivity in colon cancer cells was shown for the induction of two proapoptotic proteins, namely caveolin-1 and the tumor-suppressor gene Krüppel-like factor-4 (KLF-4). beta-CDODA-Me but not alpha-CDODA-Me induced caveolin-1 in SW480 colon cancer cells, whereas caveolin-1 was induced by both compounds in HT-29 and HCT-15 colon cancer cells. The CDODA-Me isomers induced KLF-4 mRNA levels in HT-29 and SW480 cells but had minimal effects on KLF-4 expression in HCT-15 cells. These induced responses were inhibited by cotreatment with a PPARgamma antagonist. This shows for the first time that PPARgamma agonists derived from glycyrrhetinic acid induced cell-dependent caveolin-1 and KLF-4 expression through receptor-dependent pathways.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.