1
|
Wang B, Li D, Ilnytskyy Y, Khachigian LM, Zhong N, Rodriguez-Juarez R, Kovalchuk I, Kovalchuk O. A Positive Feedback DNA-PK/MYT1L-CXCR1-ERK1/2 Proliferative Signaling Loop in Glioblastoma. Int J Mol Sci 2025; 26:4398. [PMID: 40362634 PMCID: PMC12072392 DOI: 10.3390/ijms26094398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Glioblastoma is the most common primary brain tumor in adults. Our previous studies revealed a functional interplay of myelin transcription factor 1-like (MYT1L) with the DNA-dependent protein kinase (DNA-PK) in the regulation of p21 transcription. However, the contributing role of this functional interplay in glioblastoma remains largely unknown. Here, we used cell lines with normal DNA-PK (HEK293 and M059K) or deficient DNA-PK (M059J) as a model system to demonstrate the importance of the DNA-PK-dependent activation of MYT1L in controlling the transcription of CXC chemokine receptor 1 (CXCR1) in a positive-feedback proliferative signaling loop in glioblastoma with numerous conventional techniques. In normal DNA-PK cells, MYT1L acted as an oncogene by promoting cell proliferation, inhibiting apoptosis, and shortening a cell cycle S phase. However, in DNA-PK-deficient cells, MYT1L functioned as a tumor suppressor by inhibiting cell proliferation and inducing a G1 arrest. The enforced expression of MYT1L promoted CXCR1 transcription in DNA-PK-normal cells but attenuated transcription in DNA-PK-deficient cells. Bioinformatics analysis predicted a MYT1L-binding sequence at the CXCR1 promoter. The functional dependence of MYT1L on DNA-PK in CXCR1 transcription was validated by luciferase assay. Although the expression of CXCR1 was lower in M059J cells as compared to M059K cells, it was higher than in normal brain tissue. The CXCR1 ligands interleukin 8 (IL-8) and GRO protein alpha (GROα) expressed in M059J and M059K cells may signal through the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway that can be blocked by CXCR1 siRNA. Our findings demonstrate the existence of a positive feedback DNA-PK/MYT1L-CXCR1-ERK1/2 proliferation loop in glioblastoma cells that may represent a pharmacological target loop for therapeutic intervention.
Collapse
Affiliation(s)
- Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Nuanying Zhong
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Rocio Rodriguez-Juarez
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| |
Collapse
|
2
|
All-trans retinoic acid and protein kinase C α/β1 inhibitor combined treatment targets cancer stem cells and impairs breast tumor progression. Sci Rep 2021; 11:6044. [PMID: 33723318 PMCID: PMC7961031 DOI: 10.1038/s41598-021-85344-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 02/28/2021] [Indexed: 01/08/2023] Open
Abstract
Breast cancer is the leading cause of cancer death among women worldwide. Blocking a single signaling pathway is often an ineffective therapy, especially in the case of aggressive or drug-resistant tumors. Since we have previously described the mechanism involved in the crosstalk between Retinoic Acid system and protein kinase C (PKC) pathway, the rationale of our study was to evaluate the effect of combining all-trans-retinoic acid (ATRA) with a classical PCK inhibitor (Gö6976) in preclinical settings. Employing hormone-independent mammary cancer models, Gö6976 and ATRA combined treatment induced a synergistic reduction in proliferative potential that correlated with an increased apoptosis and RARs modulation towards an anti-oncogenic profile. Combined treatment also impairs growth, self-renewal and clonogenicity potential of cancer stem cells and reduced tumor growth, metastatic spread and cancer stem cells frequency in vivo. An in-silico analysis of “Kaplan–Meier plotter” database indicated that low PKCα together with high RARα mRNA expression is a favorable prognosis factor for hormone-independent breast cancer patients. Here we demonstrate that a classical PKC inhibitor potentiates ATRA antitumor effects also targeting cancer stem cells growth, self-renewal and frequency.
Collapse
|
3
|
Castellanos-Esparza YC, Wu S, Huang L, Buquet C, Shen R, Sanchez-Gonzalez B, García Latorre EA, Boyer O, Varin R, Jiménez-Zamudio LA, Janin A, Vannier JP, Li H, Lu H. Synergistic promoting effects of pentoxifylline and simvastatin on the apoptosis of triple-negative MDA-MB-231 breast cancer cells. Int J Oncol 2018; 52:1246-1254. [PMID: 29436616 DOI: 10.3892/ijo.2018.4272] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/14/2017] [Indexed: 11/06/2022] Open
Abstract
Pentoxifylline (PTX), a xanthine family molecule and simvastatin (SIM), an anti-hypercholesterolemic agent, have recently been considered as sensitizers to chemotherapy and radiotherapy. The present in vitro study evaluated their antitumor synergistic effects on MDA‑MB‑231 breast cancer cells characterized by the triple‑negative phenotype (TNP). The anti-proliferative effects of these two agents were evaluated by MTT and clonogenic assays. Cell cycle progression was examined using propidium iodide staining. Apoptosis was investigated by Annexin V labeling, and by examining caspase 3 activity and DNA fragmentation. Autophagic vesicles and reactive oxygen species (ROS) levels were monitored by flow cytometry. Western blot analysis was performed to evaluate molecular targets. Our results revealed that when used alone, PTX and SIM exerted antitumor effects. Nevertheless, used in combination, the inhibition of cell proliferation was synergistically superior (80% vs 42%) than that observed following treatment with each agent alone after 48 h. PTX alone (0.5 mM) induced both apoptosis (25%) and autophagy (25%); however, when used in combination with SIM (0.5 µM), the balance between these processes was disrupted and the cells underwent apoptosis (>65%) as opposed to autophagy (<13%). This imbalance was associated with an increase in ERK1/2 and AKT activation, but not with an increase in mTOR phosphorylation, and with the suppression of the NF-κB pathway. In addition, in the cells treated with both agents, almost 78% of the cells were arrested at the G0/G1 phase and lost their colony-forming ability (38±5%) compared to the cells treated with PTX alone (115±5%). On the whole, these results suggest that the induction of autophagy may be a protective mechanism preventing MDA‑MB‑231 cancer cell death. The combined use of PTX and SIM may drive dormant autophagic cancer cells to undergo apoptosis and thus this may be a novel treatment strategy for breast cancer characterized by the TNP.
Collapse
Affiliation(s)
- Yessica Cristina Castellanos-Esparza
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Shuang Wu
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Limin Huang
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Catherine Buquet
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Rong Shen
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Berenice Sanchez-Gonzalez
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Ethel Awilda García Latorre
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Olivier Boyer
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Remi Varin
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Luis Antonio Jiménez-Zamudio
- Immunochemistry Laboratory I, Immunology Department, National School of Biological Sciences, National Polytechnic Institute, Mexico City 11340, Mexico
| | - Anne Janin
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| | - Jean-Pierre Vannier
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - Hong Li
- National Institute of Health and Medical Research, Unit 1234/Rouen University, Faculty of Medicine and Pharmacy, 76183 Rouen, France
| | - He Lu
- National Institute of Health and Medical Research, Medical Research Unit S-1165/Paris Diderot University, University Institute of Hematology, Saint-Louis Hospital, 75010 Paris, France
| |
Collapse
|
4
|
Kona SL, Shrestha A, Yi X, Joseph S, Barona HM, Martinez-Ceballos E. RARβ2-dependent signaling represses neuronal differentiation in mouse ES cells. Differentiation 2017; 98:55-61. [PMID: 29154149 PMCID: PMC5726922 DOI: 10.1016/j.diff.2017.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/30/2017] [Accepted: 11/08/2017] [Indexed: 01/03/2023]
Abstract
Embryonic Stem (ES) cells are pluripotent cells that can be induced to differentiate into cells of all three lineages: mesoderm, endoderm, and ectoderm. In culture, ES cells can be differentiated into mature neurons by treatment with Retinoic Acid (RA) and this effect is mediated mainly through the activation of the RA nuclear receptors (RAR α, β, and γ), and their isoforms. However, little is known about the role played by specific RAR types on ES cell differentiation. Here, we found that treatment of ES cells with AC55649, an RARβ2 agonist, increased endodermal marker gene expression. On the other hand, we found that the inhibition of RARβ with 5μM LE135, together with RA treatment, increased the efficiency of mouse ES cell differentiation into neurons by more than 4-fold as compared to cells treated with RA only. Finally, we performed proteomic analyses on ES cells treated with RA vs RA plus AC55649 in order to identify the signaling pathways activated by the RARβ agonist. Our proteomic analyses using antibody microarrays indicated that proteins such as p38 and AKT were upregulated in cells treated with RA plus the agonist, as compared to cells treated with RA alone. Our results indicate that RARβ may function as a repressor of neuronal differentiation through the activation of major cell signaling pathways, and that the pharmacological inhibition of this nuclear receptor may constitute a novel method to increase the efficiency of ES to neuronal differentiation in culture.
Collapse
Affiliation(s)
- Sri L Kona
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Amita Shrestha
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Xiaoping Yi
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Serenthia Joseph
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Humberto Munoz Barona
- Department of Physics and Mathematics, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Eduardo Martinez-Ceballos
- Department of Biological Sciences and Chemistry, Southern University and A&M College, Baton Rouge, LA 70813, USA.
| |
Collapse
|
5
|
Sn- and Ge- triorganometallics exert different cytotoxicity and modulation of migration in triple-negative breast cancer cell line MDA-MB-231. Toxicol Lett 2017; 279:16-21. [DOI: 10.1016/j.toxlet.2017.07.879] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/07/2017] [Accepted: 07/09/2017] [Indexed: 01/28/2023]
|
6
|
Lin G, Zhu S, Wu Y, Song C, Wang W, Zhang Y, Chen YL, He Z. ω-3 free fatty acids and all-trans retinoic acid synergistically induce growth inhibition of three subtypes of breast cancer cell lines. Sci Rep 2017; 7:2929. [PMID: 28592877 PMCID: PMC5462805 DOI: 10.1038/s41598-017-03231-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
All-trans retinoic acid (ATRA), one of vitamin A derivatives, shows greater growth inhibition of breast cancer cell for ER-positive than ER-negative cells, while triple negative breast cancer cell such as MDA-MB-231 cell is poorly responsive to ATRA treatment. In this study, we found that combination of ω-3 free fatty acids (ω-3 FFAs) and ATRA exhibited synergistic inhibition of cell growth in three subtypes (ER+ MCF7, HER2+ SK-BR-3, Triple negative HCC1806 and MDA-MB-231 cells) of human breast cancer cell lines. The combined treatment of ω-3 FFAs and ATRA resulted in cell cycle arrest. ω-3 FFAs combined with ATRA synergistically provoked cell apoptosis via the caspase signals but not p53. These findings suggest that combined chemotherapy of ω-3 FFAs with ATRA is beneficial for improvement of ATRA sensitivity in breast cancer cells.
Collapse
Affiliation(s)
- Guangxiao Lin
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Shenglong Zhu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Yikuan Wu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Ci Song
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Wanjing Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Yuan Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Yue-Lei Chen
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P.R. China.
| | - Zhao He
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China.
- School of Medicine, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
7
|
Complementary PTM Profiling of Drug Response in Human Gastric Carcinoma by Immunoaffinity and IMAC Methods with Total Proteome Analysis. Proteomes 2015; 3:160-183. [PMID: 28248267 PMCID: PMC5217380 DOI: 10.3390/proteomes3030160] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 01/14/2023] Open
Abstract
Gaining insight into normal cellular signaling and disease biology is a critical goal of proteomic analyses. The ability to perform these studies successfully to extract the maximum value and discovery of biologically relevant candidate biomarkers is therefore of primary importance. Many successful studies in the past have focused on total proteome analysis (changes at the protein level) combined with phosphorylation analysis by metal affinity enrichment (changes at the PTM level). Here, we use the gastric carcinoma cell line MKN-45 treated with the c-Met inhibitor SU11274 and PKC inhibitor staurosporine to investigate the most efficient and most comprehensive strategies for both total protein and PTM analysis. Under the conditions used, total protein analysis yielded few changes in response to either compound, while analysis of phosphorylation identified thousands of sites that changed differentially between the two treatments. Both metal affinity and antibody-based enrichments were used to assess phosphopeptide changes, and the data generated by the two methods was largely complementary (non-overlapping). Label-free quantitation of peptide peak abundances was used to accurately determine fold-changes between control and treated samples. Protein interaction network analysis allowed the data to be placed in a biologically relevant context, and follow-up validation of selected findings confirmed the accuracy of the proteomic data. Together, this study provides a framework for start-to-finish proteomic analysis of any experimental system under investigation to maximize the value of the proteomic study and yield the best chance for uncovering actionable target candidates.
Collapse
|
8
|
Huang B, Xiao E, Huang M. MEK/ERK pathway is positively involved in hypoxia-induced vasculogenic mimicry formation in hepatocellular carcinoma which is regulated negatively by protein kinase A. Med Oncol 2014; 32:408. [PMID: 25487444 DOI: 10.1007/s12032-014-0408-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 11/24/2014] [Indexed: 11/24/2022]
Abstract
The aim of present investigation is to explore the molecular mechanisms of vasculogenic mimicry (VM) induced by hypoxia. Hepatocellular carcinoma cell lines were treated with CoCl2, and the VM-related parameters were assayed by real-time qPCR, Western blotting and immunofluorescence. Matrigel tube structure was also detected. We demonstrated that the expression of pMEK, MEK, pERK1/2 and ERK1/2 had a positive correlation with VM induced by hypoxia in MHCC97H while HepG2 signified VM under normoxia condition. PD98059 was negatively while epidermal growth factor positively participated in the increased tubes and area of VM. At the meaning time, the increased VM-related genes VE-cadherin, MMP2, MMP9, EphA2 and LAMC2 in hypoxia group were down-regulated by PD98059 in a dose-dependent manner. Furthermore, we elucidated that PKA, but not PKC, mediated the MEK/ERK pathway in a negative manner in VM. In conclusion, MEK/ERK pathway is positively involved in VM in hepatocellular carcinoma cell line, which was mediated by PKA negatively.
Collapse
Affiliation(s)
- Bin Huang
- Radiology Department, The Second Xiangya Hospital of Central South University, No. 139, Renmin Road, Changsha, 410008, China
| | | | | |
Collapse
|
9
|
Retinoids and breast cancer: from basic studies to the clinic and back again. Cancer Treat Rev 2014; 40:739-49. [PMID: 24480385 DOI: 10.1016/j.ctrv.2014.01.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/09/2014] [Accepted: 01/10/2014] [Indexed: 11/23/2022]
Abstract
All-trans retinoic acid (ATRA) is the most important active metabolite of vitamin A controlling segmentation in the developing organism and the homeostasis of various tissues in the adult. ATRA as well as natural and synthetic derivatives, collectively known as retinoids, are also promising agents in the treatment and chemoprevention of different types of neoplasia including breast cancer. The major aim of the present article is to review the basic knowledge acquired on the anti-tumor activity of classic retinoids, like ATRA, in mammary tumors, focusing on the underlying cellular and molecular mechanisms and the determinants of retinoid sensitivity/resistance. In the first part, an analysis of the large number of pre-clinical studies available is provided, stressing the point that this has resulted in a limited number of clinical trials. This is followed by an overview of the knowledge acquired on the role played by the retinoid nuclear receptors in the anti-tumor responses triggered by retinoids. The body of the article emphasizes the potential of ATRA and derivatives in modulating and in being influenced by some of the most relevant cellular pathways involved in the growth and progression of breast cancer. We review the studies centering on the cross-talk between retinoids and some of the growth-factor pathways which control the homeostasis of the mammary tumor cell. In addition, we consider the cross-talk with relevant intra-cellular second messenger pathways. The information provided lays the foundation for the development of rational and retinoid-based therapeutic strategies to be used for the management of breast cancer.
Collapse
|
10
|
Ierano C, Basseville A, To KKW, Zhan Z, Robey RW, Wilkerson J, Bates SE, Scala S. Histone deacetylase inhibitors induce CXCR4 mRNA but antagonize CXCR4 migration. Cancer Biol Ther 2012. [PMID: 23192271 DOI: 10.4161/cbt.22957] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The stromal cell-derived factor-1α SDF-1α (CXCL12)/CXCR4 axis has been linked to poor prognosis in some cancers. As histone deacetylase inhibitors (HDIs) exert antitumor effects by targeting proteins affecting cell migration, we sought to evaluate the effects of the HDIs apicidin, vorinostat, entinostat (MS-275) and romidepsin on the expression and function of CXCR4 in human cancer cell lines. After treatment with romidepsin, CXCR4 mRNA expression increased 12-fold in UOK121 renal cancer cells, 16-fold in H460 non-small cell cancer cells and 4-fold in SF295 glioma cells; treatment with other HDIs yielded similar effects. CXCR4 induction was not observed in MCF7 breast cancer cells or SW620 colon cancer cells. To evaluate the corresponding functional increase, the effect of CXCR4 ligand, CXCL12, on ERK1/2, STAT3 and c-SRC activation and cell migration was examined in UOK121, SF295 and H460 cells. Alone, the HDIs increased pERK1/2, while reducing pSTAT-3 and pSRC. Following CXCL12 exposure, pERK1/2 induction was maintained, but STAT3 and SRC phosphorylation was impaired. These findings resulted in reduced basal and CXCL12-mediated cell migration. In conclusion, HDIs upregulated CXCR4 mRNA expression but impaired CXCL12-dependent signaling cascades through STAT3 and c-SRC, suggesting a potential role for HDIs in delaying or preventing metastatic processes in solid tumors.
Collapse
Affiliation(s)
- Caterina Ierano
- Medical Oncology Branch, National Cancer Institute; Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Zhan Y, Zhang Y, Liu C, Zhang J, Smith WW, Wang N, Chen Y, Zheng L, He L. A Novel Taspine Derivative, HMQ1611, Inhibits Breast Cancer Cell Growth via Estrogen Receptor α and EGF Receptor Signaling Pathways. Cancer Prev Res (Phila) 2012; 5:864-73. [DOI: 10.1158/1940-6207.capr-11-0575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
12
|
Schobert R, Seibt S, Mahal K, Ahmad A, Biersack B, Effenberger-Neidnicht K, Padhye S, Sarkar FH, Mueller T. Cancer Selective Metallocenedicarboxylates of the Fungal Cytotoxin Illudin M. J Med Chem 2011; 54:6177-82. [DOI: 10.1021/jm200359n] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Rainer Schobert
- Organic Chemistry Laboratory, University Bayreuth, 95447 Bayreuth, Germany
| | - Sebastian Seibt
- Organic Chemistry Laboratory, University Bayreuth, 95447 Bayreuth, Germany
| | - Katharina Mahal
- Organic Chemistry Laboratory, University Bayreuth, 95447 Bayreuth, Germany
| | - Aamir Ahmad
- Department for Pathology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, 95447 Bayreuth, Germany
| | | | - Subhash Padhye
- Department for Pathology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
- Center for Drug Design and Molecular Medicine, Department of Environmental Sciences, University of Pune, Pune 411 007, India
| | - Fazlul H. Sarkar
- Department for Pathology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| | - Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| |
Collapse
|
13
|
Pettersson F, Yau C, Dobocan MC, Culjkovic-Kraljacic B, Retrouvey H, Puckett R, Flores LM, Krop IE, Rousseau C, Cocolakis E, Borden KLB, Benz CC, Miller WH. Ribavirin treatment effects on breast cancers overexpressing eIF4E, a biomarker with prognostic specificity for luminal B-type breast cancer. Clin Cancer Res 2011; 17:2874-84. [PMID: 21415224 DOI: 10.1158/1078-0432.ccr-10-2334] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We have evaluated the eukaryotic translation initiation factor 4E (eIF4E) as a potential biomarker and therapeutic target in breast cancer. eIF4E facilitates nuclear export and translation of specific, growth-stimulatory mRNAs and is frequently overexpressed in cancer. EXPERIMENTAL DESIGN Breast cancer cells were treated with ribavirin, an inhibitor of eIF4E, and effects on cell proliferation and on known mRNA targets of eIF4E were determined. eIF4E expression was assessed, at the mRNA and protein level, in breast cancer cell lines and in skin biopsies from patients with metastatic disease. Additionally, pooled microarray data from 621 adjuvant untreated, node-negative breast cancers were analyzed for eIF4E expression levels and correlation with distant metastasis-free survival (DMFS), overall and within each intrinsic breast cancer subtype. RESULTS At clinically relevant concentrations, ribavirin reduced cell proliferation and suppressed clonogenic potential, correlating with reduced mRNA export and protein expression of important eIF4E targets. This effect was suppressed by knockdown of eIF4E. Although eIF4E expression is elevated in all breast cancer cell lines, variability in ribavirin responsiveness was observed, indicating that other factors contribute to an eIF4E-dependent phenotype. Assessment of the prognostic value of high eIF4E mRNA in patient tumors found that significant discrimination between good and poor outcome groups was observed only in luminal B cases, suggesting that a specific molecular profile may predict response to eIF4E-targeted therapy. CONCLUSIONS Inhibition of eIF4E is a potential breast cancer therapeutic strategy that may be especially promising against specific molecular subtypes and in metastatic as well as primary tumors.
Collapse
MESH Headings
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/classification
- Breast Neoplasms/diagnosis
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Carcinoma/classification
- Carcinoma/diagnosis
- Carcinoma/drug therapy
- Carcinoma/genetics
- Cell Line, Tumor
- Cells, Cultured
- Eukaryotic Initiation Factor-4E/antagonists & inhibitors
- Eukaryotic Initiation Factor-4E/genetics
- Eukaryotic Initiation Factor-4E/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Knockdown Techniques
- Humans
- Mammary Glands, Human/metabolism
- Mammary Glands, Human/pathology
- Organ Specificity/genetics
- Prognosis
- RNA, Small Interfering/pharmacology
- Ribavirin/pharmacology
- Ribavirin/therapeutic use
- Up-Regulation/drug effects
- Up-Regulation/genetics
Collapse
Affiliation(s)
- Filippa Pettersson
- Lady Davis Institute & Segal Cancer Centre of the Jewish General Hospital, McGill University, Montréal, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Soto-Guzman A, Navarro-Tito N, Castro-Sanchez L, Martinez-Orozco R, Salazar EP. Oleic acid promotes MMP-9 secretion and invasion in breast cancer cells. Clin Exp Metastasis 2010; 27:505-15. [PMID: 20617371 DOI: 10.1007/s10585-010-9340-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 06/24/2010] [Indexed: 01/20/2023]
Abstract
Epidemiological and animal studies suggest an association between dietary fatty acids and an increase risk of developing breast cancer. Obesity, which is characterized by hyperlipidemia and an elevation of circulating free fatty acids (FFAs), is also associated with enhanced cancer risk. In breast cancer cells, the FFA oleic acid (OA) induces migration, proliferation, prolong survival, invasion, an increase in cellular Ca(2+) concentration, MEK1/2, ERK1/2, FAK and Src activation. However, the role of OA on MMP-9 secretion and invasion has not been studied in detail. We demonstrate here that stimulation of MDA-MB-231 breast cancer cells with 200 μM OA induces an increase on MMP-9 secretion through a PKC, Src, and EGFR-dependent pathway, as revealed by gelatin zymography assays. Furthermore, microtubule network mediates MMP-9 secretion induced by OA. In contrast, OA does not induce an increase on MMP-9 secretion in MCF10A cells, whereas it does not induce MMP-9 secretion in MCF12A mammary non-tumorigenic epithelial cells. In addition, OA induces invasion through an EGFR, Gi/Go proteins, MMPs, PKC and Src-dependent pathway, but it is not able to promote invasion in non-invasive MCF-7 breast cancer cells. In summary, our findings demonstrate that OA promotes an increase on MMP-9 secretion and invasion through a PKC, Src, and EGFR-dependent pathway in breast cancer cells.
Collapse
Affiliation(s)
- Adriana Soto-Guzman
- Departamento de Biologia Celular, Cinvestav-IPN, San Pedro Zacatenco, Mexico, DF, Mexico
| | | | | | | | | |
Collapse
|
15
|
Afrasiabi E, Hietamäki M, Viitanen T, Sukumaran P, Bergelin N, Törnquist K. Expression and significance of HERG (KCNH2) potassium channels in the regulation of MDA-MB-435S melanoma cell proliferation and migration. Cell Signal 2010; 22:57-64. [DOI: 10.1016/j.cellsig.2009.09.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Accepted: 09/09/2009] [Indexed: 01/15/2023]
|
16
|
Lønne GK, Masoumi KC, Lennartsson J, Larsson C. Protein kinase Cdelta supports survival of MDA-MB-231 breast cancer cells by suppressing the ERK1/2 pathway. J Biol Chem 2009; 284:33456-65. [PMID: 19833733 DOI: 10.1074/jbc.m109.036186] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mechanisms that mediate apoptosis resistance are attractive therapeutic targets for cancer. Protein kinase Cdelta (PKCdelta) is considered a pro-apoptotic factor in many cell types. In breast cancer, however, it has shown both pro-survival and pro-apoptotic effects. Here, we report for the first time that down-regulation of PKCdelta per se leads to apoptosis of MDA-MB-231 cells. Inhibition of MEK1/2 by either PD98059 or U0126 suppressed the induction of apoptosis of PKCdelta-depleted MDA-MB-231 cells but did not support survival of MCF-7 or MDA-MB-468 cells. Basal ERK1/2 phosphorylation was substantially higher in MDA-MB-231 cells than in the other cell lines. PKCdelta depletion led to even higher ERK1/2 phosphorylation levels and also to lower expression levels of the ERK1/2 phosphatase MKP3. Depletion of MKP3 led to apoptosis and higher levels of ERK1/2 phosphorylation, suggesting that this may be a mechanism mediating the effect of PKCdelta down-regulation. However, PKCdelta silencing also induced increased MEK1/2 phosphorylation, indicating that PKCdelta regulates ERK1/2 phosphorylation both upstream and downstream. Moreover, PKCdelta silencing led to increased levels of the E3 ubiquitin ligase Nedd4, which is a potential regulator of MKP3, because down-regulation led to increased MKP3 levels. Our results highlight PKCdelta as a potential target for therapy of breast cancers with high activity of the ERK1/2 pathway.
Collapse
Affiliation(s)
- Gry Kalstad Lønne
- Center for Molecular Pathology, Department of Laboratory Medicine, Lund University, and Malmö University Hospital, Sweden
| | | | | | | |
Collapse
|
17
|
Zheng B, Han M, Bernier M, Zhang XH, Meng F, Miao SB, He M, Zhao XM, Wen JK. Krüppel-like factor 4 inhibits proliferation by platelet-derived growth factor receptor beta-mediated, not by retinoic acid receptor alpha-mediated, phosphatidylinositol 3-kinase and ERK signaling in vascular smooth muscle cells. J Biol Chem 2009; 284:22773-85. [PMID: 19531492 DOI: 10.1074/jbc.m109.026989] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Proliferation inhibition of vascular smooth muscle cells (VSMCs) is governed by the activity of a transcription factor network. Krüppel-like factor 4 (Klf4), retinoic acid receptor (RAR alpha), and platelet-derived growth factor receptor (PDGFR) are expressed in VSMCs and are components of such a network. However, the relationship among them in the regulation of VSMC proliferation remains unknown. Here, we investigated the mechanisms whereby Klf4 mediates the growth inhibitory effects in VSMCs through RAR alpha and PDGFR beta. We demonstrated that Klf4 directly binds to the 5' regulatory region of RAR alpha, down-regulates RAR alpha expression, and specifically inhibits RAR alpha-mediated phosphatidylinositol 3-kinase (PI3K) and ERK signaling in cultured VSMCs induced by the synthetic retinoid Am80. Of particular interest, Klf4 inhibits RAR alpha and PDGFR beta expression while blocking PI3K and ERK signaling induced by Am80 and PDGF-BB, respectively. The anti-proliferative effects of Klf4 on neointimal formation depend largely on PDGFR-mediated PI3K signaling without involvement of the RAR alpha-activated signaling pathways. These findings provide a novel mechanism for signal suppression and growth inhibitory effects of Klf4 in VSMCs. Moreover, the results of this study suggest that Klf4 is one of the key mediators of retinoid actions in VSMCs.
Collapse
Affiliation(s)
- Bin Zheng
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Zhongshan East Road, Shijiazhuang 050017, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Fukumura M, Ando H, Hirai Y, Toriizuka K, Ida Y, Kuchino Y. Achyranthoside H methyl ester, a novel oleanolic acid saponin derivative from Achyranthes fauriei roots, induces apoptosis in human breast cancer MCF-7 and MDA-MB-453 cells via a caspase activation pathway. J Nat Med 2009; 63:181-8. [DOI: 10.1007/s11418-008-0311-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Accepted: 12/05/2008] [Indexed: 11/28/2022]
|
19
|
Afrasiabi E, Ahlgren J, Bergelin N, Törnquist K. Phorbol 12-myristate 13-acetate inhibits FRO anaplastic human thyroid cancer cell proliferation by inducing cell cycle arrest in G1/S phase: evidence for an effect mediated by PKCdelta. Mol Cell Endocrinol 2008; 292:26-35. [PMID: 18541361 DOI: 10.1016/j.mce.2008.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 04/30/2008] [Accepted: 04/30/2008] [Indexed: 01/06/2023]
Abstract
Phorbol 12-myristate 13-acetate (PMA) is known to affect a variety of cellular processes, including cell proliferation, differentiation, and migration. PMA has been shown to promote antiproliferative and antimigratory effects in many types of cancer cells. Our findings show that PMA induced a strong antiproliferative effect in two anaplastic (FRO and ARO) and one follicular (ML-1) thyroid cancer cell lines, and increased the fraction of FRO cells in G1 phase of the cell cycle. The fractions in the S and G2 phases were decreased. Moreover, PMA evoked a significant increase in the levels of the cell cycle regulators p21Waf1/Cip1 and p27Kip1. The levels of cyclin D3 and the cyclin-dependent kinases cdk4 and cdk6 decreased, as did the phosphorylation of the Rb-protein. PMA did not induce apoptosis. PMA stimulated the translocation of protein kinase C (PKC) alpha, betaI and delta isoforms to the cell membrane. PKCdelta small interfering RNA attenuated the PMA-induced antiproliferative effect and prevented the upregulation of p21Waf1/Cip1 and p27Kip1. Prolonged stimulation with PMA decreased the phosphorylation of mitogen-activated protein (MAP) kinase. PMA also decreased the phosphorylation of Akt and evoked a biphasic change in the phosphorylation of the forkhead box class-O protein (FOXO): an increase in phosphorylation, followed by a dephosphorylation. In addition, PMA inhibited FRO, ARO and ML-1 cell migration toward serum. The inactive phorbol ester analog 4alpha-phorbol and the diacylglycerol analog 1,2-dioctanoyl-sn-glycerol were without an effect on proliferation and migration. The results indicate that PMA is an effective inhibitor of thyroid cancer cell proliferation and migration by a mechanism involving PKC-MAP kinase/Akt and FOXO signaling.
Collapse
Affiliation(s)
- Emad Afrasiabi
- Department of Biology, Abo Akademi University, BioCity, Artillerigatan 6, 20520 Turku, Finland
| | | | | | | |
Collapse
|
20
|
Nelson AM, Zhao W, Gilliland KL, Zaenglein AL, Liu W, Thiboutot DM. Neutrophil gelatinase-associated lipocalin mediates 13-cis retinoic acid-induced apoptosis of human sebaceous gland cells. J Clin Invest 2008; 118:1468-78. [PMID: 18317594 DOI: 10.1172/jci33869] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Accepted: 01/16/2008] [Indexed: 01/13/2023] Open
Abstract
13-cis retinoic acid (13-cis RA; also known as isotretinoin) is the most potent agent available for treatment of acne. It is known that the drug induces apoptosis in cells cultured from human sebaceous glands, but its mechanism of action has not been determined. In this study, skin biopsies were taken from 7 patients with acne prior to and at 1 week of treatment with 13-cis RA. TUNEL staining confirmed that 13-cis RA induced apoptosis in sebaceous glands. Transcriptional profiling of patient skin and cultured human sebaceous gland cells (SEB-1 sebocytes) indicated that lipocalin 2 was among the genes most highly upregulated by 13-cis RA. Lipocalin 2 encodes neutrophil gelatinase-associated lipocalin (NGAL), which functions in innate immune defense and induces apoptosis of murine B lymphocytes. Increased immunolocalization of NGAL was noted in patients' sebaceous glands following treatment with 13-cis RA, and recombinant NGAL induced apoptosis in SEB-1 sebocytes. Furthermore, apoptosis in response to 13-cis RA was inhibited in the presence of siRNA to lipocalin 2. These data indicate that NGAL mediates the apoptotic effect of 13-cis RA and suggest that agents that selectively induce NGAL expression in sebaceous glands might represent therapeutic alternatives to the use of 13-cis RA to treat individuals with acne.
Collapse
|
21
|
Delaune A, Corbière C, Benjelloun FD, Legrand E, Vannier JP, Ripoll C, Vasse M. Promyelocytic leukemia-nuclear body formation is an early event leading to retinoic acid-induced differentiation of neuroblastoma cells. J Neurochem 2007; 104:89-99. [PMID: 17986232 DOI: 10.1111/j.1471-4159.2007.05019.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neuroblastoma is one of the most common cancers in children. Neuroblastoma differentiation is linked to the presence of the promyelocytic leukemia (PML) protein. Retinoic acid, a powerful differentiation-inducer in vitro, is a potent agent for the treatment of neuroblastoma. Using two different human neuroblastoma cell lines, SH-SY5Y and LA-N-5, we show here that PML protein leads to the formation of nuclear bodies (PML-NB) after only 1 h of retinoic acid treatment and that this formation is mediated by the extracellular signal-regulated kinase (ERK) pathway. Inhibition of protein kinase C also leads to formation of PML-NB via the ERK pathway. Both sumoylation and phosphorylation of PML in an ERK-dependent pathway are also required for formation of PML-NB. Finally, we show that PML-NB formation in neuroblastoma cells is associated with neurite outgrowth. These results support the proposal that the formation of PML-NB is correlated with the differentiation of neuroblastoma cells.
Collapse
Affiliation(s)
- A Delaune
- Groupe de recherche MERCI & IHURBM, Faculté de médecine et pharmacie, Rouen, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Papageorgiou E, Pitulis N, Msaouel P, Lembessis P, Koutsilieris M. The non-genomic crosstalk between PPAR-gamma ligands and ERK1/2 in cancer cell lines. Expert Opin Ther Targets 2007; 11:1071-85. [PMID: 17665979 DOI: 10.1517/14728222.11.8.1071] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peroxisome proliferator activated receptors (PPARs) are members of the nuclear receptor superfamily acting as transcription factors. PPAR-gamma, one of the three PPAR subtypes, is expressed in many malignant and non-malignant cells and tissues. PPAR-gamma ligands influence cancer biology via both genomic as well as non-genomic events. The non-genomic action of PPAR-gamma ligands, including the activation of MAPK signaling pathways, is under intense investigation. In the presence of PPAR-gamma ligands, a rapid phosphorylation of ERK1/2 is observed in many cancer cell lines. Activated ERK1/2 elicits rapid, non-genomic cellular effects and can directly repress PPAR-gamma transcriptional activity by phosphorylation. This paper reviews the interrelation of PPAR-gamma ligands and activated ERK1/2, in relation to their antineoplastic actions in cancer cell lines, which may offer the potential for improved anticancer therapies.
Collapse
Affiliation(s)
- Efstathia Papageorgiou
- National & Kapodistrian University of Athens, Department of Experimental Physiology, Medical School, Goudi-Athens, Greece
| | | | | | | | | |
Collapse
|
23
|
Ryu JY, Whang J, Park H, Im JY, Kim J, Ahn MY, Lee J, Kim HS, Lee BM, Yoo SD, Kwack SJ, Oh JH, Park KL, Han SY, Kim SH. Di(2-ethylhexyl) phthalate induces apoptosis through peroxisome proliferators-activated receptor-gamma and ERK 1/2 activation in testis of Sprague-Dawley rats. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2007; 70:1296-1303. [PMID: 17654247 DOI: 10.1080/15287390701432160] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a well-known hepatic and reproductive toxicant whose toxicity may be mediated by peroxisome proliferators-activated receptor (PPAR). This study examined the effects of DEHP on the expression of PPAR-regulated genes involved in testicular cells apoptosis. Sprague-Dawley male rats were treated orally with 250, 500, or 750 mg/kg/d DEHP for 28 d, while control rats were given corn oil. The levels of cell cycle regulators (pRb, cyclins, CDKs, and p21) and apoptosis-related proteins were analyzed by Western blot analysis. The role of PPAR-gamma (PPAR-gamma), class B scavenger receptor type 1 (SR-B1), and ERK1/2 was further studied to examine the signaling pathway for DEHP-induced apoptosis. Results showed that the levels of pRB, cyclin D, CDK2, cyclin E, and CDK4 were significantly lower in rats given 500 and 750 mg/kg/d DEHP, while levels of p21 were significantly higher in rat testes. Dose-dependent increases in PPAR-gamma and RXRalpha proteins were observed in testes after DEHP exposure, while there was a significant decrease in RXRgamma protein levels. In addition to PPAR-gamma, DEHP also significantly increased SR-B1 mRNA and phosphorylated ERK1/2 protein levels. Furthermore, DEHP treatment induced pro-caspase-3 and cleavage of its substrate protein, poly(ADP-ribose) polymerase (PARP), in a dose-dependent manner. Data suggest that DEHP exposure may induce the expression of apoptosis-related genes in testes through induction of PPAR-gamma and activation of the ERK1/2 pathway.
Collapse
Affiliation(s)
- Ju Young Ryu
- College of Pharmacy, Pusan National University, Pusan, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Son SH, Yu E, Ahn Y, Choi EK, Lee H, Choi J. Retinoic acid attenuates promyelocytic leukemia protein-induced cell death in breast cancer cells by activation of the ubiquitin–proteasome pathway. Cancer Lett 2007; 247:213-23. [PMID: 16740359 DOI: 10.1016/j.canlet.2006.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 03/28/2006] [Accepted: 04/24/2006] [Indexed: 02/07/2023]
Abstract
All-trans-retinoic acid and the tumor suppressor promyelocytic leukemia protein (PML) are potent regulators of the growth of cancer cells. This study investigates the individual and combined effects of PML, when overexpressed by the recombinant PML adenovirus, and all-trans-retinoic acid on the proliferation of human estrogen-receptor negative SKBR-3 and estrogen-receptor positive MCF-7 breast cancer cell lines. All-trans-retinoic acid caused a significant degree of cell death in SKBR-3 cells and MCF-7 cells, and PML elicited a similar incidence of or slightly more cell death in MCF-7 cells. Dual-treated cells displayed significantly less cell death than did single-treated cells in the same cell line. We concluded that PML and all-trans-retinoic acid cause cell death by different pathways: PML activates ERK1/2, p38 MAPK, and p21; arrests the cell cycle; and later causes cell death; and all-trans-retinoic acid activates proteasome function, caspase cleavage, and apoptosis. The combined use of all-trans-retinoic acid and PML gene therapy may not be the best treatment for patients with cancer, because the ubiquitinylation of PML and its subsequent proteasome-dependent degradation by retinoic acids occur before overexpressed PML exhibits tumor-suppressive activity.
Collapse
Affiliation(s)
- Se-Hee Son
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
25
|
Garattini E, Gianni' M, Terao M. Cytodifferentiation by retinoids, a novel therapeutic option in oncology: rational combinations with other therapeutic agents. VITAMINS AND HORMONES 2007; 75:301-54. [PMID: 17368321 DOI: 10.1016/s0083-6729(06)75012-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Retinoic acid (RA) and derivatives are promising antineoplastic agents endowed with both therapeutic and chemopreventive potential. Although the treatment of acute promyelocytic leukemia with all-trans retinoic acid is an outstanding example, the full potential of retinoids in oncology has not yet been explored and a more generalized use of these compounds is not yet a reality. One way to enhance the therapeutic and chemopreventive activity of RA and derivatives is to identify rational combinations between these compounds and other pharmacological agents. This is now possible given the information available on the biochemical and molecular mechanisms underlying the biological activity of retinoids. At the cellular level, the antileukemia and anticancer activity of retinoids is the result of three main actions, cytodifferentiation, growth inhibition, and apoptosis. Cytodifferentiation is a particularly attractive modality of treatment and differentiating agents promise to be less toxic and more specific than conventional chemotherapy. This is the result of the fact that cytotoxicity is not the primary aim of differentiation therapy. At the molecular level, retinoids act through the activation of nuclear retinoic acid receptor-dependent and -independent pathways. The cellular pathways and molecular networks relevant for retinoid activity are modulated by a panoply of other intracellular and extracellular pathways that may be targeted by known drugs and other experimental therapeutics. This chapter aims to summarize and critically discuss the available knowledge in the field.
Collapse
Affiliation(s)
- Enrico Garattini
- Laboratorio di Biologia Molecolare, Centro Catullo e Daniela Borgomainerio, Istituto di Ricerche Farmacologiche Mario Negri, via Eritrea 62, 20157 Milano, Italy
| | | | | |
Collapse
|
26
|
Abstract
Peroxisome proliferator-activated receptors gamma (PPAR?) is a transcriptional factor belonging to the ligand-activated nuclear receptor superfamily. PPAR? is highly expressed in adipose tissue and has a dominant regulatory role in adipocyte differentiation. In humans, PPAR? is expressed in multiple tissues such as the breast, colon, lung, ovary, and placenta. In addition to adipogenic and anti-inflammatory effects, PPAR? activation has been shown to be anti-proliferative by virtue of its differentiation-promoting effect, suggesting that activation of PPAR? may be useful in slowing or arresting the proliferation of de-differentiated tumor cells. A number of PPAR? ligands, such as natural prostaglandins and synthetic anti-diabetic thiazolidinediones (TZDs), have been identified. The discovery of PPAR? agonists has enabled the elucidation of the mechanisms involved in the multiple effects of PPAR? on the inhibition of tumor cell growth. The importance of this transcription factor in physiology and pathophysiology has stimulated much research in this field. This review describes structural features of PPAR?, mechanisms of PPAR? gene transcription, and recent developments in the discovery of its biological functions on growth inhibition of lung tumors. Prospects for future research leading to new therapies for lung cancer are also discussed.
Collapse
Affiliation(s)
- Ming-Yue Li
- Department of Surgery, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | | | | | | |
Collapse
|
27
|
Koike K, Fujii T, Nakamura AM, Yokoyama G, Yamana H, Kuwano M, Shirouzu K. Activation of protein kinase C delta induces growth arrest in NPA thyroid cancer cells through extracellular signal-regulated kinase mitogen-activated protein kinase. Thyroid 2006; 16:333-41. [PMID: 16646678 DOI: 10.1089/thy.2006.16.333] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Protein kinase C (PKC) is a family of serine-threonine kinases that regulate many cell processes. To study the role of PKCdelta in thyroid cancer cells, we used a replication-deficient adenovirus (PKCdeltaAdV), to tightly control PKCdelta expression. In NPA cells, activation of wild-type (WT) PKCdelta with phorbol 12-myristate 13-acetate (PMA) induced an arrest in cell growth at G(1) phase, which was itself inhibited by the PKCdelta inhibitor rottlerin. Furthermore, overexpression of a dominant negative PKCdelta did not induce G(1) arrest. These findings strongly suggested that PKCdelta induced cell growth arrest in NPA cells. We investigated the mechanism of G1 arrest by examining G(1)-related proteins and mitogen-activated protein kinase (MAPK) by Western blotting. After activation of WTPKCdelta with PMA, cyclin E expression and retinoblastoma protein (Rb) phosphorylation decreased; the expression of p27(Kip1) increased and the phosphorylation of extracellular signal-regulated kinase (ERK) MAPK decreased. These results indicated that the activation of PKCdelta induced cell growth arrest in NPA cells, through an ERK MAPK-p27(Kip1)-cyclin E-pRb pathway. PKCdelta may therefore be an effective molecular target for novel therapy in thyroid cancer.
Collapse
Affiliation(s)
- Kenta Koike
- Department of Surgery, Kurume University School of Medicine, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Li M, Lee TW, Yim APC, Mok TSK, Chen GG. Apoptosis induced by troglitazone is both peroxisome proliferator-activated receptor-γ- and ERK-dependent in human non-small lung cancer cells. J Cell Physiol 2006; 209:428-38. [PMID: 16883598 DOI: 10.1002/jcp.20738] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The role of the peroxisome proliferator-activated receptor-gamma (PPARgamma) in cell differentiation, cell-cycle arrest, and apoptosis has attracted increasing attention. We have recently demonstrated that PPARgamma ligands-troglitazone (TGZ) induced apoptosis in lung cancer cells. In this report, we further studied the role of ERK1/2 in lung cancer cells treated by TGZ. The result demonstrated that TGZ induced PPARgamma and ERK1/2 accumulation in the nucleus, in which the co-localization of both proteins was found. The activation of ERK1/2 resulted in apoptosis via a mitochondrial pathway. Both PPARgamma siRNA and U0126, a specific inhibitor of ERK1/2, were able to block these effects of TGZ, suggesting that apoptosis induced by TGZ was PPARgamma and ERK1/2 dependent. Inhibition of ERK1/2 by U0126 also led to a significant decrease in the level of PPARgamma, indicating a positive cross-talk between PPARgamma and ERK1/2 or an auto-regulatory feedback mechanism to amplify the effect of ERK1/2 on cell growth arrest and apoptosis. In addition to ERK1/2, TGZ also activated Akt. Interestingly, inhibition of ERK1/2 prevented the activation of Akt whereas the suppression of Akt had no effect on ERK1/2, suggesting that Akt was not necessary for TGZ-PPARgamma-ERK pathway. However, the inhibition of Akt promoted the release of cytochrome c, suggesting the activation of Akt may have a negative effect on apoptosis induced by TGZ. In conclusion, our study has demonstrated that TGZ, a synthetic PPARgamma ligand, induced apoptosis in NCI-H23 lung cancer cells via a mitochondrial pathway and this pathway was PPARgamma and ERK1/2 dependent.
Collapse
Affiliation(s)
- Mingyue Li
- Department of Surgery, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | | | | | | | | |
Collapse
|
29
|
Mashour GA, Drissel SN, Frahm S, Farassati F, Martuza RL, Mautner VF, Kindler-Röhrborn A, Kurtz A. Differential modulation of malignant peripheral nerve sheath tumor growth by omega-3 and omega-6 fatty acids. Oncogene 2005; 24:2367-74. [PMID: 15735744 DOI: 10.1038/sj.onc.1208425] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common genetic disorder of the nervous system resulting in neurofibromas and malignant peripheral nerve sheath tumors (MPNST). In this study, we report the modulation of murine and human MPNST cell growth by the fatty acids docosahexaenoic acid (DHA) and arachidonic acid (AA). DHA demonstrated a tendency to stimulate cell growth at low doses and induce apoptosis at high doses, paralleled by the activation of ERK and caspase-3. Furthermore, high-dose DHA reversed the stimulation of MPNST cell growth by a number of growth factors suggested to have a pathogenic effect in NF1 and inhibited MPNST growth in vivo. AA was found to have a reciprocal activity in vitro, stimulating MPNST cell growth at comparable concentrations and reducing DHA activation of ERK. These findings introduce fatty acids as a possible regulator of MPNST development in NF1 patients.
Collapse
Affiliation(s)
- George A Mashour
- Molecular Neurosurgery Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Gossé F, Guyot S, Roussi S, Lobstein A, Fischer B, Seiler N, Raul F. Chemopreventive properties of apple procyanidins on human colon cancer-derived metastatic SW620 cells and in a rat model of colon carcinogenesis. Carcinogenesis 2005; 26:1291-5. [PMID: 15790589 DOI: 10.1093/carcin/bgi074] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Apples contain several classes of polyphenols: monomers (catechins, epicatechins) and oligomers/polymers, such as the procyanidins. Our aim was (i) to study anti-proliferative mechanisms on human metastatic colon carcinoma (SW620 cells) of apple polyphenol fractions (monomers or procyanidins) and (ii) to evaluate their anti-carcinogenic properties in vivo. Two polyphenol-enriched fractions were isolated from apples. Fraction non-procyanidins contained 73% phenolic monomers and no procyanidins, while fraction procyanidins contained 78% procyanidins and no monomers. Inhibition of SW620 cell growth was only observed with fraction P (IC50 = 45 microg/ml). After a 24-h exposure of cells to fraction P, protein kinase C activity was inhibited by 70% and a significant increase in extracellular signal-regulated kinases 1 and 2 and c-jun N-terminal kinases expression was observed together with the down-regulation of polyamine biosynthesis and the activation of caspase-3. Colon carcinogenesis was induced in rats by intraperitoneal injections of azoxymethane, once a week for 2 weeks. Seven days after the last injection, Wistar rats received fraction P (0.01%) dissolved in drinking water. After 6 weeks of treatment, the colon of rats receiving procyanidins showed a significant (P < 0.01) reduction of the number of preneoplastic lesions when compared with controls receiving water. The total number of hyperproliferative crypts and of aberrant crypt foci was reduced by 50% in rats receiving 0.01% apple procyanidins in their drinking water. Our results show that apple procyanidins alter intracellular signaling pathways, polyamine biosynthesis and trigger apoptosis in tumor cells. These compounds antagonize cancer promotion in vivo. In contrast with absorbable drugs, these natural, non toxic, dietary constituents reach the colon where they are able to exert their antitumor effects.
Collapse
Affiliation(s)
- Francine Gossé
- Laboratoire d'Oncologie Nutritionnelle, Université Louis Pasteur EA 3430, Institut de Recherche contre les Cancers de l'Appareil Digestif (IRCAD), Strasbourg, France
| | | | | | | | | | | | | |
Collapse
|
31
|
Wu W, Pew T, Zou M, Pang D, Conzen SD. Glucocorticoid receptor-induced MAPK phosphatase-1 (MPK-1) expression inhibits paclitaxel-associated MAPK activation and contributes to breast cancer cell survival. J Biol Chem 2004; 280:4117-24. [PMID: 15590693 DOI: 10.1074/jbc.m411200200] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucocorticoid receptor (GR) activation has recently been shown to inhibit apoptosis in breast epithelial cells. We have previously described a group of genes that is rapidly up-regulated in these cells following dexamethasone (Dex) treatment. In an effort to dissect the mechanisms of GR-mediated breast epithelial cell survival, we now examine the molecular events downstream of GR activation. Here we show that GR activation leads to both the rapid induction of MAPK phosphatase-1 (MKP-1) mRNA and its sustained expression. Induction of the MKP-1 protein in the MCF10A-Myc and MDA-MB-231 breast epithelial cell lines was also seen. Paclitaxel treatment resulted in MAPK activation and apoptosis of MDA-MB-231 breast cancer cells, and both processes were inhibited by Dex pretreatment. Furthermore, induction of MKP-1 correlated with the inhibition of extracellular signal-regulated kinase (ERK1/2) and c-Jun N-terminal kinase (JNK) activity, whereas p38 activity was minimally affected. Blocking Dex-induced MKP-1 induction using small interfering RNA increased ERK1/2 and JNK phosphorylation and decreased cell survival. ERK1/2 and JNK inactivation was associated with Ets-like transcription factor-1 (ELK-1) dephosphorylation. To explore the gene expression changes that occur downstream of ELK-1 dephosphorylation, we used a combination of temporal gene expression data and promoter element analyses. This approach revealed a previously unrecognized transcriptional target of ELK-1, the human tissue plasminogen activator (tPA). We verified the predicted ELK-1--> tPA transcriptional regulatory relationship using a luciferase reporter assay. We conclude that GR-mediated MAPK inactivation contributes to cell survival and that the potential transcriptional targets of this inhibition can be identified from large scale gene array analysis.
Collapse
MESH Headings
- Amino Acid Motifs
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis
- Blotting, Northern
- Blotting, Western
- Breast/metabolism
- Cell Cycle Proteins/metabolism
- Cell Cycle Proteins/physiology
- Cell Line, Tumor
- Cell Survival
- DNA-Binding Proteins/biosynthesis
- Down-Regulation
- Dual Specificity Phosphatase 1
- Enzyme Activation
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Humans
- Immediate-Early Proteins/metabolism
- Immediate-Early Proteins/physiology
- JNK Mitogen-Activated Protein Kinases/biosynthesis
- JNK Mitogen-Activated Protein Kinases/metabolism
- Luciferases/metabolism
- MAP Kinase Kinase 4
- MAP Kinase Signaling System
- Mitogen-Activated Protein Kinase 1/biosynthesis
- Mitogen-Activated Protein Kinase 3/biosynthesis
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Mitogen-Activated Protein Kinases/metabolism
- Models, Biological
- Oligonucleotide Array Sequence Analysis
- Paclitaxel/pharmacology
- Phosphoprotein Phosphatases/metabolism
- Phosphoprotein Phosphatases/physiology
- Phosphorylation
- Protein Binding
- Protein Phosphatase 1
- Protein Tyrosine Phosphatases/metabolism
- Protein Tyrosine Phosphatases/physiology
- Proto-Oncogene Proteins/biosynthesis
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Receptors, Glucocorticoid/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
- Tissue Plasminogen Activator/metabolism
- Transcription Factors/biosynthesis
- Transcription, Genetic
- Transfection
- ets-Domain Protein Elk-1
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine and the Committee on Cancer Biology, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | |
Collapse
|