51
|
Ganesh S, Koser ML, Cyr WA, Chopda GR, Tao J, Shui X, Ying B, Chen D, Pandya P, Chipumuro E, Siddiquee Z, Craig K, Lai C, Dudek H, Monga SP, Wang W, Brown BD, Abrams MT. Direct Pharmacological Inhibition of β-Catenin by RNA Interference in Tumors of Diverse Origin. Mol Cancer Ther 2016; 15:2143-54. [PMID: 27390343 DOI: 10.1158/1535-7163.mct-16-0309] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 06/29/2016] [Indexed: 01/30/2023]
Abstract
The Wnt/β-catenin pathway is among the most frequently altered signaling networks in human cancers. Despite decades of preclinical and clinical research, efficient therapeutic targeting of Wnt/β-catenin has been elusive. RNA interference (RNAi) technology silences genes at the mRNA level and therefore can be applied to previously undruggable targets. Lipid nanoparticles (LNP) represent an elegant solution for the delivery of RNAi-triggering oligonucleotides to disease-relevant tissues, but have been mostly restricted to applications in the liver. In this study, we systematically tuned the composition of a prototype LNP to enable tumor-selective delivery of a Dicer-substrate siRNA (DsiRNA) targeting CTNNB1, the gene encoding β-catenin. This formulation, termed EnCore-R, demonstrated pharmacodynamic activity in subcutaneous human tumor xenografts, orthotopic patient-derived xenograft (PDX) tumors, disseminated hematopoietic tumors, genetically induced primary liver tumors, metastatic colorectal tumors, and murine metastatic melanoma. DsiRNA delivery was homogeneous in tumor sections, selective over normal liver and independent of apolipoprotein-E binding. Significant tumor growth inhibition was achieved in Wnt-dependent colorectal and hepatocellular carcinoma models, but not in Wnt-independent tumors. Finally, no evidence of accelerated blood clearance or sustained liver transaminase elevation was observed after repeated dosing in nonhuman primates. These data support further investigation to gain mechanistic insight, optimize dose regimens, and identify efficacious combinations with standard-of-care therapeutics. Mol Cancer Ther; 15(9); 2143-54. ©2016 AACR.
Collapse
Affiliation(s)
- Shanthi Ganesh
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts.
| | | | - Wendy A Cyr
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | | | - Junyan Tao
- University of Pittsburgh Medical Center, Pittsburgh, Pittsburgh
| | - Xue Shui
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Bo Ying
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Dongyu Chen
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Purva Pandya
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | | | | | - Kevin Craig
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Chengjung Lai
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Henryk Dudek
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | | | - Weimin Wang
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Bob D Brown
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| | - Marc T Abrams
- Dicerna Pharmaceuticals, Inc., Cambridge, Massachusetts
| |
Collapse
|
52
|
Chen Y, Zhang Y. Functional and mechanistic analysis of telomerase: An antitumor drug target. Pharmacol Ther 2016; 163:24-47. [PMID: 27118336 DOI: 10.1016/j.pharmthera.2016.03.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/29/2016] [Indexed: 01/26/2023]
|
53
|
Boudjadi S, Carrier JC, Groulx JF, Beaulieu JF. Integrin α1β1 expression is controlled by c-MYC in colorectal cancer cells. Oncogene 2016; 35:1671-1678. [PMID: 26096932 PMCID: PMC4820680 DOI: 10.1038/onc.2015.231] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 04/15/2015] [Accepted: 05/10/2015] [Indexed: 12/13/2022]
Abstract
The α1β1 collagen receptor is only present in a few epithelial cell types. In the intestine, it is specifically expressed in proliferating crypt cells. This integrin has been reported to be involved in various cancers where it mediates the downstream activation of the Ras/ERK proliferative pathway. We have recently shown that integrin α1β1 is present in two-thirds of colon adenocarcinomas, but the mechanism by which ITGA1 expression is regulated is not known. DNA methylation, involved in ITGA1 repression during megakaryocyte differentiation, is not the mechanism of ITGA1 regulation in colorectal cancer cells. Our in silico analysis of the ITGA1 promoter revealed two response elements for MYC, an oncogenic factor known to regulate cancer cell proliferation, invasion and migration. In situ, the expressions of both MYC and ITGA1 are localized in the lower crypt of the normal colon and correlate in 72% of the 65 analyzed colorectal cancers. MYC pharmacological inhibition or downregulation of expression with short hairpin RNA in HT29, T84 and SW480 cells resulted in reduced ITGA1 expression at both the transcript and protein levels. Chromatin immunoprecipitation assays revealed that MYC was bound to the chromatin region of the ITGA1 proximal promoter, whereas MYC overexpression enhanced ITGA1 promoter activity that was reduced with MAD co-transfection or by the disruption of the response elements. We concluded that MYC is a key regulating factor for the control of ITGA1 expression.
Collapse
Affiliation(s)
- S Boudjadi
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - J C Carrier
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - J-F Groulx
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - J-F Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
54
|
Hellerbrand C, Massoumi R. Cylindromatosis-A Protective Molecule against Liver Diseases. Med Res Rev 2016; 36:342-59. [DOI: 10.1002/med.21381] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/12/2015] [Accepted: 11/17/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Claus Hellerbrand
- Department of Internal Medicine I; University Hospital Regensburg; 93053 Regensburg Germany
| | - Ramin Massoumi
- Department of Laboratory Medicine, Medicon Village; Lund University; 22381 Lund Sweden
| |
Collapse
|
55
|
Wang F, Remke M, Bhat K, Wong ET, Zhou S, Ramaswamy V, Dubuc A, Fonkem E, Salem S, Zhang H, Hsieh TC, O'Rourke ST, Wu L, Li DW, Hawkins C, Kohane IS, Wu JM, Wu M, Taylor MD, Wu E. A microRNA-1280/JAG2 network comprises a novel biological target in high-risk medulloblastoma. Oncotarget 2015; 6:2709-24. [PMID: 25576913 PMCID: PMC4413612 DOI: 10.18632/oncotarget.2779] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 11/19/2014] [Indexed: 01/23/2023] Open
Abstract
Over-expression of PDGF receptors (PDGFRs) has been previously implicated in high-risk medulloblastoma (MB) pathogenesis. However, the exact biological functions of PDGFRα and PDGFRβ signaling in MB biology remain poorly understood. Here, we report the subgroup specific expression of PDGFRα and PDGFRβ and their associated biological pathways in MB tumors. c-MYC, a downstream target of PDGFRβ but not PDGFRα, is involved in PDGFRβ signaling associated with cell proliferation, cell death, and invasion. Concurrent inhibition of PDGFRβ and c-MYC blocks MB cell proliferation and migration synergistically. Integrated analysis of miRNA and miRNA targets regulated by both PDGFRβ and c-MYC reveals that increased expression of JAG2, a target of miR-1280, is associated with high metastatic dissemination at diagnosis and a poor outcome in MB patients. Our study may resolve the controversy on the role of PDGFRs in MB and unveils JAG2 as a key downstream effector of a PDGFRβ-driven signaling cascade and a potential therapeutic target.
Collapse
Affiliation(s)
- Fengfei Wang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Marc Remke
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Kruttika Bhat
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Eric T Wong
- Brain Tumor Center & Neuro-Oncology Unit, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Shuang Zhou
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Vijay Ramaswamy
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Adrian Dubuc
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Ekokobe Fonkem
- Scott & White Neuroscience Institute, Texas A & M Health Science Center, Temple, TX 76508, USA
| | - Saeed Salem
- Department of Computer Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Hongbing Zhang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100073, China
| | - Tze-Chen Hsieh
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Stephen T O'Rourke
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, Shands Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - David W Li
- Department of Ophthalmology & Visual Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cynthia Hawkins
- Division of Pathology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Isaac S Kohane
- Informatics Program, Children's Hospital Boston, Harvard Medical School, Boston 02115, MA, USA
| | - Joseph M Wu
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Min Wu
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, ND 58202, USA
| | - Michael D Taylor
- Arthur and Sonia Labatt Brain Tumor Research Centre, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
56
|
Liu Q, Nguyen E, Døskeland S, Ségal-Bendirdjian É. cAMP-Dependent Protein Kinase A (PKA)-Mediated c-Myc Degradation Is Dependent on the Relative Proportion of PKA-I and PKA-II Isozymes. Mol Pharmacol 2015; 88:469-76. [PMID: 26104548 DOI: 10.1124/mol.115.097915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/23/2015] [Indexed: 02/14/2025] Open
Abstract
The transcription factor c-Myc regulates numerous target genes that are important for multiple cellular processes such as cell growth and differentiation. It is commonly deregulated in leukemia. Acute promyelocytic leukemia (APL) is characterized by a blockade of granulocytic differentiation at the promyelocyte stage. Despite the great success of all-trans retinoic acid (ATRA)-based therapy, which results in a clinical remission by inducing promyelocyte maturation, a significant number of patients relapse due to the development of ATRA resistance. A significant role has been ascribed to the cAMP/cAMP-dependent protein kinase A (PKA) signaling pathway in retinoid treatment since PKA activation is able to restore differentiation in some ATRA-resistant cells and eradicate leukemia-initiating cells in vivo. In this study, using NB4 APL cell variants resistant to ATRA-induced differentiation, we reveal distinct functional roles of the two PKA isozymes, PKA type I (PKA-I) and PKA-type II (PKA-II), on the steady-state level of c-Myc protein, providing a likely mechanism by which cAMP-elevating agents can restore differentiation in ATRA maturation-resistant APL cells. Therefore, both the inhibition of c-Myc activity and the PKA-I/PKA-II ratio should be taken into account if cAMP-based therapy is considered in the clinical management of APL.
Collapse
Affiliation(s)
- Qingyuan Liu
- Institut National de la Santé et de la Recherche Médicale UMR-S 1007, Homéostasie Cellulaire et Cancer, Université Paris-Descartes, Paris Sorbonne Cité, Paris, France (Q.L., E.N., E.S.-B.); and Department of Biomedicine, University of Bergen, Bergen, Norway (S.D.)
| | - Eric Nguyen
- Institut National de la Santé et de la Recherche Médicale UMR-S 1007, Homéostasie Cellulaire et Cancer, Université Paris-Descartes, Paris Sorbonne Cité, Paris, France (Q.L., E.N., E.S.-B.); and Department of Biomedicine, University of Bergen, Bergen, Norway (S.D.)
| | - Stein Døskeland
- Institut National de la Santé et de la Recherche Médicale UMR-S 1007, Homéostasie Cellulaire et Cancer, Université Paris-Descartes, Paris Sorbonne Cité, Paris, France (Q.L., E.N., E.S.-B.); and Department of Biomedicine, University of Bergen, Bergen, Norway (S.D.)
| | - Évelyne Ségal-Bendirdjian
- Institut National de la Santé et de la Recherche Médicale UMR-S 1007, Homéostasie Cellulaire et Cancer, Université Paris-Descartes, Paris Sorbonne Cité, Paris, France (Q.L., E.N., E.S.-B.); and Department of Biomedicine, University of Bergen, Bergen, Norway (S.D.)
| |
Collapse
|
57
|
Schrecengost RS, Keller SN, Schiewer MJ, Knudsen KE, Smith CD. Downregulation of Critical Oncogenes by the Selective SK2 Inhibitor ABC294640 Hinders Prostate Cancer Progression. Mol Cancer Res 2015; 13:1591-601. [PMID: 26271487 DOI: 10.1158/1541-7786.mcr-14-0626] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 07/30/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED The bioactive sphingolipid sphingosine-1-phosphate (S1P) drives several hallmark processes of cancer, making the enzymes that synthesize S1P, that is, sphingosine kinase 1 and 2 (SK1 and SK2), important molecular targets for cancer drug development. ABC294640 is a first-in-class SK2 small-molecule inhibitor that effectively inhibits cancer cell growth in vitro and in vivo. Given that AR and Myc are two of the most widely implicated oncogenes in prostate cancer, and that sphingolipids affect signaling by both proteins, the therapeutic potential for using ABC294640 in the treatment of prostate cancer was evaluated. This study demonstrates that ABC294640 abrogates signaling pathways requisite for prostate cancer growth and proliferation. Key findings validate that ABC294640 treatment of early-stage and advanced prostate cancer models downregulate Myc and AR expression and activity. This corresponds with significant inhibition of growth, proliferation, and cell-cycle progression. Finally, oral administration of ABC294640 was found to dramatically impede xenograft tumor growth. Together, these pre-clinical findings support the hypotheses that SK2 activity is required for prostate cancer function and that ABC294640 represents a new pharmacological agent for treatment of early stage and aggressive prostate cancer. IMPLICATIONS Sphingosine kinase inhibition disrupts multiple oncogenic signaling pathways that are deregulated in prostate cancer.
Collapse
Affiliation(s)
| | - Staci N Keller
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania
| | - Matthew J Schiewer
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Karen E Knudsen
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Urology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania. Department of Radiation Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Charles D Smith
- Apogee Biotechnology Corporation, Hummelstown, Pennsylvania.
| |
Collapse
|
58
|
Reyes-González JM, Armaiz-Peña GN, Mangala LS, Valiyeva F, Ivan C, Pradeep S, Echevarría-Vargas IM, Rivera-Reyes A, Sood AK, Vivas-Mejía PE. Targeting c-MYC in Platinum-Resistant Ovarian Cancer. Mol Cancer Ther 2015; 14:2260-9. [PMID: 26227489 DOI: 10.1158/1535-7163.mct-14-0801] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 07/19/2015] [Indexed: 11/16/2022]
Abstract
The purpose of this study was to investigate the molecular and therapeutic effects of siRNA-mediated c-MYC silencing in cisplatin-resistant ovarian cancer. Statistical analysis of patient's data extracted from The Cancer Genome Atlas (TCGA) portal showed that the disease-free (DFS) and the overall (OS) survival were decreased in ovarian cancer patients with high c-MYC mRNA levels. Furthermore, analysis of a panel of ovarian cancer cell lines showed that c-MYC protein levels were higher in cisplatin-resistant cells when compared with their cisplatin-sensitive counterparts. In vitro cell viability, growth, cell-cycle progression, and apoptosis, as well as in vivo therapeutic effectiveness in murine xenograft models, were also assessed following siRNA-mediated c-MYC silencing in cisplatin-resistant ovarian cancer cells. Significant inhibition of cell growth and viability, cell-cycle arrest, and activation of apoptosis were observed upon siRNA-mediated c-MYC depletion. In addition, single weekly doses of c-MYC-siRNA incorporated into 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG-2000)-based nanoliposomes resulted in significant reduction in tumor growth. These findings identify c-MYC as a potential therapeutic target for ovarian cancers expressing high levels of this oncoprotein.
Collapse
Affiliation(s)
- Jeyshka M Reyes-González
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Guillermo N Armaiz-Peña
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lingegowda S Mangala
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fatma Valiyeva
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| | - Cristina Ivan
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunila Pradeep
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Adrian Rivera-Reyes
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pablo E Vivas-Mejía
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico. University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico.
| |
Collapse
|
59
|
Inhibition of c-Myc by 10058-F4 induces growth arrest and chemosensitivity in pancreatic ductal adenocarcinoma. Biomed Pharmacother 2015. [DOI: 10.1016/j.biopha.2015.05.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
60
|
Wang X, Zhou P, Sun X, Zheng J, Wei G, Zhang L, Wang H, Yao J, Lu S, Jia P. Acidified bile acids increase hTERT expression via c-myc activation in human gastric cancer cells. Oncol Rep 2015; 33:3038-44. [PMID: 25873431 DOI: 10.3892/or.2015.3908] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/24/2015] [Indexed: 11/06/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) is upregulated in most cancer cell types as well in immortalized cells. The underlying mechanism for such upregulation, however, remains largely unknown. We report here that bile acids under acidified media increase hTERT expression via c-myc activation in primary human gastric cancer cell lines. Human gastric cancer MKN28, MGC803 and SGC7901 cells were treated with 100 µM deoxycholic acid (DCA) or chenodeoxycholic acid (CDCA) with or without acidified media in the presence or absence of the c-myc inhibitor 10058-F4 for 24 h. hTERT and c-myc protein levels were determined by western blot analysis. hTERT and c-myc mRNA levels were determined by RT-PCR. The promoter activities of hTERT and c-myc transcription were determined using promoter reporter luciferase assays for both. Telomerase enzyme activity was analyzed by stretch PCR. hTERT mRNA and protein levels were significantly increased by bile acids in acidified media and were accompanied with enhanced telomerase activity. No changes were found at a pH of 7.0 or with acidified media alone. Similarly, the mRNA and protein levels of c-myc were also increased by bile acids in acidified media but not at a pH of 7.0 or with acidified media alone. Importantly, pharmacologic inhibition of c-myc using 10058-F4 prevented hTERT induction by DCA or CDCA in gastric cancer cells under acidic conditions. Bile acids (DCA and CDCA) under acidic conditions increased hTERT expression in human gastric cancer cells by activation of c-myc transcription. This suggests that acidified bile acids may promote tumorigenesis and affect cell ageing via telomerase activation.
Collapse
Affiliation(s)
- Xiaolong Wang
- Department of General Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Peihua Zhou
- Department of General Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xuejun Sun
- Department of General Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianbao Zheng
- Department of General Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Guangbing Wei
- Department of General Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Li Zhang
- Department of General Surgery, The Second Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hui Wang
- Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Jianfeng Yao
- Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Shaoying Lu
- Department of General Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Pengbo Jia
- The First People's Hospital of Xianyang City, Xianyang, Shaanxi 712000, P.R. China
| |
Collapse
|
61
|
Pan D, Kim B, Hu G, Gupta DS, Senpan A, Yang X, Schmieder A, Swain C, Wickline SA, Tomasson MH, Lanza GM. A strategy for combating melanoma with oncogenic c-Myc inhibitors and targeted nanotherapy. Nanomedicine (Lond) 2015; 10:241-51. [PMID: 25600969 PMCID: PMC4665613 DOI: 10.2217/nnm.14.101] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS The activity of the transcription factor c-Myc is dependent upon heterodimerization with Max to control target gene transcription. Small-molecule inhibitors of c-Myc-Max have exhibited low potency and poor water solubility and are therefore unsuitable for in vivo application. We hypothesized that a nanomedicine approach incorporating a cryptic c-Myc inhibitor prodrug could be delivered and enzymatically released in order to effectively inhibit melanoma. MATERIALS & METHODS An Sn-2 lipase-labile Myc inhibitor prodrug was synthesized and included in two αvβ3-targeted nanoparticle platforms (20 and 200 nm). The inherent antiproliferate potency was compared with the lipid-free compound using human and mouse melanoma cell lines. RESULTS & CONCLUSION These data demonstrate for the first time a successful nanodelivery of c-Myc inhibitors and their potential use to prevent melanoma.
Collapse
Affiliation(s)
- Dipanjan Pan
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Biomedical Research Center, Office 3304, 3rd Floor, Mills Breast Cancer Institute & Carle Foundation Hospital, 502 N. Busey, Urbana, IL 61801, USA
| | - Benjamin Kim
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Grace Hu
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Deepti Sood Gupta
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Angana Senpan
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Xiaoxia Yang
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Anne Schmieder
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Corban Swain
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Samuel A Wickline
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Michael H Tomasson
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| | - Gregory M Lanza
- Department of Medicine, Washington University School of Medicine, 4320 Forest Park Avenue, Saint Louis, MO 63108, USA
| |
Collapse
|
62
|
Wang J, Ma X, Jones HM, Chan LLY, Song F, Zhang W, Bae-Jump VL, Zhou C. Evaluation of the antitumor effects of c-Myc-Max heterodimerization inhibitor 100258-F4 in ovarian cancer cells. J Transl Med 2014; 12:226. [PMID: 25143136 PMCID: PMC4160551 DOI: 10.1186/s12967-014-0226-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 08/05/2014] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian carcinoma is the most lethal gynecological cancer due to its silent onset and recurrence with resistance to chemotherapy. Overexpression of oncogene c-Myc is one of the most frequently encountered events present in ovarian carcinoma. Disrupting the function of c-Myc and its downstream target genes is a promising strategy for cancer therapy. Our objective was to evaluate the potential effects of small-molecule c-Myc inhibitor, 10058-F4, on ovarian carcinoma cells and the underlying mechanisms by which 10058-F4 exerts its actions. Using MTT assay, colony formation, flow cytometry and Annexin V FITC assays, we found that 10058-F4 significantly inhibited cell proliferation of both SKOV3 and Hey ovarian cancer cells in a dose dependent manner through induction of apoptosis and cell cycle G1 arrest. Treatment with 10058-F4 reduced cellular ATP production and ROS levels in SKOV3 and Hey cells. Consistently, primary cultures of ovarian cancer treated with 10058-F4 showed induction of caspase-3 activity and inhibition of cell proliferation in 15 of 18 cases. The response to 10058-F4 was independent the level of c-Myc protein over-expression in primary cultures of ovarian carcinoma. These novel findings suggest that the growth of ovarian cancer cells is dependent upon c-MYC activity and that targeting c-Myc-Max heterodimerization could be a potential therapeutic strategy for ovarian cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Weiyuan Zhang
- Department of Gynecological Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China.
| | | | | |
Collapse
|
63
|
Pan XN, Chen JJ, Wang LX, Xiao RZ, Liu LL, Fang ZG, Liu Q, Long ZJ, Lin DJ. Inhibition of c-Myc overcomes cytotoxic drug resistance in acute myeloid leukemia cells by promoting differentiation. PLoS One 2014; 9:e105381. [PMID: 25127121 PMCID: PMC4134294 DOI: 10.1371/journal.pone.0105381] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/22/2014] [Indexed: 01/04/2023] Open
Abstract
Nowadays, drug resistance still represents a major obstacle to successful acute myeloid leukemia (AML) treatment and the underlying mechanism is not fully elucidated. Here, we found that high expression of c-Myc was one of the cytogenetic characteristics in the drug-resistant leukemic cells. c-Myc over-expression in leukemic cells induced resistance to chemotherapeutic drugs, enhanced colony formation capacity and inhibited cell differentiation induced by all-trans retinoic acid (ATRA). Meanwhile, inhibition of c-Myc by shRNA or specific c-Myc inhibitor 10058-F4 rescued the sensitivity to cytotoxic drugs, restrained the colony formation ability and promoted differentiation. RT-PCR and western blotting analysis showed that down-regulation of C/EBPβ contributed to the poor differentiation state of leukemic cells induced by c-Myc over-expression. Importantly, over-expression of C/EBPβ could reverse c-Myc induced drug resistance. In primary AML cells, the c-Myc expression was negatively correlated with C/EBPβ. 10058-F4, displayed anti-proliferative activity and increased cellular differentiation with up-regulation of C/EBPβ in primary AML cells. Thus, our study indicated that c-Myc could be a novel target to overcome drug resistance, providing a new approach in AML therapy.
Collapse
Affiliation(s)
- Xiao-Na Pan
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Jia-Jie Chen
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Le-Xun Wang
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Ruo-Zhi Xiao
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Ling-Ling Liu
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Gang Fang
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Quentin Liu
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - Zi-Jie Long
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
- * E-mail: (ZL); (DL)
| | - Dong-Jun Lin
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Sun Yat-sen Institute of Hematology, Sun Yat-sen University, Guangzhou, China
- * E-mail: (ZL); (DL)
| |
Collapse
|
64
|
Combination of hepatocyte specific delivery and transformation dependent expression of shRNA inducing transcriptional gene silencing of c-Myc promoter in hepatocellular carcinoma cells. BMC Cancer 2014; 14:582. [PMID: 25108398 PMCID: PMC4153911 DOI: 10.1186/1471-2407-14-582] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/28/2014] [Indexed: 12/11/2022] Open
Abstract
Background A specific targeting modality for hepatocellular carcinoma (HCC) could ideally encompass a liver cell specific delivery system of a transcriptional unit that is active only in neoplastic cells. Sendai virosomes, derived from Sendai viral envelopes, home to hepatocytes based on the liver specific expression of asialoglycoprotein receptors (ASGPRs) which are recognized by the Sendai virosomal fusion (F) proteins. As reported earlier by us and other groups, transcriptional gene silencing (TGS) does not require continuous presence of the effector siRNA/shRNA molecule and is heritable, involving epigenetic modifications, leading to long term transcriptional repression. This could be advantageous over conventional gene therapy approaches, since continuous c-Myc inactivation is required to suppress hepatocarcinoma cells. Methods Exploiting such virosomal delivery, the alpha-fetoprotein (AFP) promoter, in combination with various tumour specific enhancers, was used to drive the expression of shRNA directed against ME1a1 binding site of the proto-oncogene c-Myc P2 promoter, in order to induce TGS in neoplastic liver cells. Results The dual specificity achieved by the Sendai virosomal delivery system and the promoter/enhancer guided expression ensured that the shRNA inducing TGS was active only in liver cells that had undergone malignant transformation. Our results indicate that such a bimodal therapeutic system induced specific activation of apoptosis in hepatocarcinoma cells due to heterochromatization and increased DNA methylation of the CpG islands around the target loci. Conclusions The Sendai virosomal delivery system, combined with AFP promoter/enhancer expression machinery, could serve as a generalized mechanism for the expression of genes deleterious to transformed hepatocarcinoma cells. In this system, the epigenetic suppression of c-Myc could have an added advantage for inducing cell death in the targeted cells. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-582) contains supplementary material, which is available to authorized users.
Collapse
|
65
|
Distinctive pharmacological differences between liver cancer cell lines HepG2 and Hep3B. Cytotechnology 2014; 67:1-12. [PMID: 25002206 DOI: 10.1007/s10616-014-9761-9] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 06/23/2014] [Indexed: 01/11/2023] Open
Abstract
As cellular models for in vitro liver cancer and toxicity studies, HepG2 and Hep3B are the two most frequently used liver cancer cell lines. Because of their similarities they are often treated as the same in experimental studies. However, there are many differences that have been largely over-sighted or ignored between them. In this review, we summarize the differences between HepG2 and Hep3B cell lines that can be found in the literature based on PubMed search. We particularly focus on the differential gene expression, differential drug responses (chemosensitivity, cell cycle and growth inhibition, and gene induction), signaling pathways associated with these differences, as well as the factors in governing these differences between HepG2 and Hep3B cell lines. Based on our analyses of the available data, we suggest that neither HBx nor p53 may be the crucial factor to determine the differences between HepG2 and Hep3B cell lines although HBx regulates the expression of the majority of genes that are differentially expressed between HepG2 and Hep3B. Instead, the different maturation stages in cancer development of the original specimen between HepG2 and Hep3B may be responsible for the differences between them. This review provides insight into the molecular mechanisms underlying the differences between HepG2 and Hep3B and help investigators especially the beginners in the areas of liver cancer research and drug metabolism to fully understand, and thus better use and interpret the data from these two cell lines in their studies.
Collapse
|
66
|
Müller I, Larsson K, Frenzel A, Oliynyk G, Zirath H, Prochownik EV, Westwood NJ, Henriksson MA. Targeting of the MYCN protein with small molecule c-MYC inhibitors. PLoS One 2014; 9:e97285. [PMID: 24859015 PMCID: PMC4032254 DOI: 10.1371/journal.pone.0097285] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 04/17/2014] [Indexed: 12/27/2022] Open
Abstract
Members of the MYC family are the most frequently deregulated oncogenes in human cancer and are often correlated with aggressive disease and/or poorly differentiated tumors. Since patients with MYCN-amplified neuroblastoma have a poor prognosis, targeting MYCN using small molecule inhibitors could represent a promising therapeutic approach. We have previously demonstrated that the small molecule 10058-F4, known to bind to the c-MYC bHLHZip dimerization domain and inhibiting the c-MYC/MAX interaction, also interferes with the MYCN/MAX dimerization in vitro and imparts anti-tumorigenic effects in neuroblastoma tumor models with MYCN overexpression. Our previous work also revealed that MYCN-inhibition leads to mitochondrial dysfunction resulting in accumulation of lipid droplets in neuroblastoma cells. To expand our understanding of how small molecules interfere with MYCN, we have now analyzed the direct binding of 10058-F4, as well as three of its analogs; #474, #764 and 10058-F4(7RH), one metabolite C-m/z 232, and a structurally unrelated c-MYC inhibitor 10074-G5, to the bHLHZip domain of MYCN. We also assessed their ability to induce apoptosis, neurite outgrowth and lipid accumulation in neuroblastoma cells. Interestingly, all c-MYC binding molecules tested also bind MYCN as assayed by surface plasmon resonance. Using a proximity ligation assay, we found reduced interaction between MYCN and MAX after treatment with all molecules except for the 10058-F4 metabolite C-m/z 232 and the non-binder 10058-F4(7RH). Importantly, 10074-G5 and 10058-F4 were the most efficient in inducing neuronal differentiation and lipid accumulation in MYCN-amplified neuroblastoma cells. Together our data demonstrate MYCN-binding properties for a selection of small molecules, and provide functional information that could be of importance for future development of targeted therapies against MYCN-amplified neuroblastoma.
Collapse
Affiliation(s)
- Inga Müller
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Larsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Frenzel
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ganna Oliynyk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Hanna Zirath
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Edward V. Prochownik
- Section of Hematology/Oncology, Children's Hospital of Pittsburgh of UMPC, Pittsburgh, Pennsylvania, United States of America
| | - Nicholas J. Westwood
- School of Chemistry and Biomedical Sciences Research Complex, University of St. Andrews and EaStCHEM, St. Andrews, Fife, Scotland, United Kingdom
| | - Marie Arsenian Henriksson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
67
|
Targeting RNA polymerase I to treat MYC-driven cancer. Oncogene 2014; 34:403-12. [PMID: 24608428 DOI: 10.1038/onc.2014.13] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 02/06/2023]
Abstract
The MYC oncoprotein and transcription factor is dysregulated in a majority of human cancers and is considered a major driver of the malignant phenotype. As such, developing drugs for effective inhibition of MYC in a manner selective to malignancies is a 'holy grail' of transcription factor-based cancer therapy. Recent advances in elucidating MYC biology in both normal cells and pathological settings were anticipated to bring inhibition of tumorigenic MYC function closer to the clinic. However, while the extensive array of cellular pathways that MYC impacts present numerous fulcrum points on which to leverage MYC's therapeutic potential, identifying the critical target(s) for MYC-specific cancer therapy has been difficult to achieve. Somewhat unexpectedly, MYC's fundamental role in regulating the 'housekeeping' process of ribosome biogenesis, one of the most ubiquitously required and conserved cell functions, may provide the Achilles' heel for therapeutically targeting MYC-driven tumors.
Collapse
|
68
|
Qu X, Shen L, Zheng Y, Cui Y, Feng Z, Liu F, Liu J. A signal transduction pathway from TGF-β1 to SKP2 via Akt1 and c-Myc and its correlation with progression in human melanoma. J Invest Dermatol 2014; 134:159-167. [PMID: 23792459 DOI: 10.1038/jid.2013.281] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 10/12/2012] [Accepted: 10/29/2012] [Indexed: 12/31/2022]
Abstract
Both SKP2 (S-phase kinase-associated protein 2) and transforming growth factor-β1 (TGF-β1) play important roles in cancer metastasis through different mechanisms: TGF-β1 via induction of epithelial-mesenchymal transition (EMT) and SKP2 via downregulating p27(kip1). Recent studies indicated that c-Myc and Akt1 were active players in metastasis. In this study we demonstrated a crosstalk between these pathways. Specifically, we found that TGF-β1 treatment increased SKP2 expression accompanied with increased phosphorylation of Akt1 and c-Myc protein accumulation during EMT. We demonstrated that Akt1 was required for TGF-β1-mediated SKP2 upregulation and that c-Myc transcription factor specifically bound to the promoter of SKP2 for its enhanced transcription. Analysis of 25 samples of normal human skin, nevi, and melanomas revealed a positive correlation between c-Myc and SKP2 accumulation. Furthermore, accumulation of SKP2 and c-Myc proteins was significantly higher in metastatic melanoma samples as compared with that in primary melanomas, which again was higher than that in normal skin or nevi. In summary, our results integrated TGF-β1 signals to SKP2 via Akt1 and c-Myc during EMT, and provided, to our knowledge, a previously unreported mechanistic molecular event for TGF-β1-induced metastasis in human melanoma.
Collapse
Affiliation(s)
- Xuan Qu
- Institute of Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China
| | - Liangliang Shen
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yang Cui
- Institute of Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China
| | - Zhihui Feng
- Center for Mitochondrial Biology and Medicine, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Feng Liu
- Department of Medicine, University of California Irvine Medical School, Irvine, California, USA; Chao Family Comprehensive Cancer Center, University of California Irvine Medical School, Irvine, California, USA.
| | - Jiankang Liu
- Institute of Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China
| |
Collapse
|
69
|
Wu Q, Chen T, Zhang Z, Liao S, Wu X, Wu J, Mei W, Chen Y, Wu W, Zeng L, Zheng W. Microwave-assisted synthesis of arene ruthenium(ii) complexes [(η6-RC6H5)Ru(m-MOPIP)Cl]Cl (R = -H and -CH3) as groove binder to c-myc G4 DNA. Dalton Trans 2014; 43:9216-25. [DOI: 10.1039/c3dt53635a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Two arene Ru(ii) complexes are prepared under microwave irradiation and display application potential as small molecule inhibitors of c-myc G4 DNA.
Collapse
Affiliation(s)
- Qiong Wu
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
| | - Tianfeng Chen
- Department of Chemistry
- Jinan University
- Guangzhou, P.R. China
| | - Zhao Zhang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
| | - Siyan Liao
- School of Pharmaceutical Sciences
- Guangzhou Medical University
- Guangzhou, P.R. China
| | - Xiaohui Wu
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
| | - Jian Wu
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
| | - Wenjie Mei
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
- Department of Chemistry
- Jinan University
| | - Yanhua Chen
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
| | - Weili Wu
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
| | - Lingli Zeng
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou, P.R. China
| | - Wenjie Zheng
- Department of Chemistry
- Jinan University
- Guangzhou, P.R. China
| |
Collapse
|
70
|
Li Z, Dong L, Dean E, Yang LV. Acidosis decreases c-Myc oncogene expression in human lymphoma cells: a role for the proton-sensing G protein-coupled receptor TDAG8. Int J Mol Sci 2013; 14:20236-55. [PMID: 24152439 PMCID: PMC3821613 DOI: 10.3390/ijms141020236] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/15/2013] [Accepted: 09/16/2013] [Indexed: 01/11/2023] Open
Abstract
Acidosis is a biochemical hallmark of the tumor microenvironment. Here, we report that acute acidosis decreases c-Myc oncogene expression in U937 human lymphoma cells. The level of c-Myc transcripts, but not mRNA or protein stability, contributes to c-Myc protein reduction under acidosis. The pH-sensing receptor TDAG8 (GPR65) is involved in acidosis-induced c-Myc downregulation. TDAG8 is expressed in U937 lymphoma cells, and the overexpression or knockdown of TDAG8 further decreases or partially rescues c-Myc expression, respectively. Acidic pH alone is insufficient to reduce c-Myc expression, as it does not decrease c-Myc in H1299 lung cancer cells expressing very low levels of pH-sensing G protein-coupled receptors (GPCRs). Instead, c-Myc is slightly increased by acidosis in H1299 cells, but this increase is completely inhibited by ectopic overexpression of TDAG8. Interestingly, TDAG8 expression is decreased by more than 50% in human lymphoma samples in comparison to non-tumorous lymph nodes and spleens, suggesting a potential tumor suppressor function of TDAG8 in lymphoma. Collectively, our results identify a novel mechanism of c-Myc regulation by acidosis in the tumor microenvironment and indicate that modulation of TDAG8 and related pH-sensing receptor pathways may be exploited as a new approach to inhibit Myc expression.
Collapse
Affiliation(s)
- Zhigang Li
- Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; E-Mails: (Z.L.); (L.D.); (E.D.)
| | - Lixue Dong
- Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; E-Mails: (Z.L.); (L.D.); (E.D.)
| | - Eric Dean
- Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; E-Mails: (Z.L.); (L.D.); (E.D.)
| | - Li V. Yang
- Department of Oncology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; E-Mails: (Z.L.); (L.D.); (E.D.)
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
- Lineberger Comprehensive Cancer Center, UNC at Chapel Hill, NC 27599, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-252-744-3419; Fax: +1-252-744-3418
| |
Collapse
|
71
|
Pannem RR, Dorn C, Ahlqvist K, Bosserhoff AK, Hellerbrand C, Massoumi R. CYLD controls c-MYC expression through the JNK-dependent signaling pathway in hepatocellular carcinoma. Carcinogenesis 2013; 35:461-8. [PMID: 24104553 DOI: 10.1093/carcin/bgt335] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Posttranslational modification of different proteins via direct ubiquitin attachment is vital for mediating various cellular processes. Cylindromatosis (CYLD), a deubiquitination enzyme, is able to cleave the polyubiquitin chains from the substrate and to regulate different signaling pathways. Loss, or reduced expression, of CYLD is observed in different types of human cancer, such as hepatocellular carcinoma (HCC). However, the molecular mechanism by which CYLD affects cancerogenesis has to date not been unveiled. The aim of the present study was to examine how CYLD regulates cellular functions and signaling pathways during hepatocancerogenesis. We found that mice lacking CYLD were highly susceptible to chemically induced liver cancer. The mechanism behind proved to be an elevated proliferation rate of hepatocytes, owing to sustained c-Jun N-terminal kinase 1 (JNK1)-mediated signaling via ubiquitination of TNF receptor-associated factor 2 and expression of c-MYC. Overexpression of wild-type CYLD in HCC cell lines prevented cell proliferation, without affecting apoptosis, adhesion and migration. A combined immunohistochemical and tissue microarray analysis of 81 human HCC tissues revealed that CYLD expression is negatively correlated with expression of proliferation markers Ki-67 and c-MYC. To conclude, we found that downregulation of CYLD induces tumor cell proliferation, consequently contributing to the aggressive growth of HCC. Our findings suggest that CYLD holds potential to serve as a marker for HCC progression, and its link to c-MYC via JNK1 may provide the foundation for new therapeutic strategies for HCC patients.
Collapse
Affiliation(s)
- Rajeswara Rao Pannem
- Department of Laboratory Medicine, Skåne University Hospital, Lund University, Malmö SE-20502, Sweden
| | | | | | | | | | | |
Collapse
|
72
|
Song A, Ye J, Zhang K, Sun L, Zhao Y, Yu H. Lentiviral vector-mediated siRNA knockdown of c-MYC: cell growth inhibition and cell cycle arrest at G2/M phase in Jijoye cells. Biochem Genet 2013; 51:603-17. [PMID: 23657834 DOI: 10.1007/s10528-013-9590-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 10/16/2012] [Indexed: 12/25/2022]
Abstract
Inhibition of c-MYC has been considered as a potential therapy for lymphoma treatment. We explored a lentiviral vector-mediated small interfering RNA (siRNA) expression vector to stably reduce c-MYC expression in B cell line Jijoye cells and investigated the effects of c-MYC downregulation on cell growth, cell cycle, and apoptosis in vitro. The expression of c-MYC mRNA and protein levels were inhibited significantly by c-MYC siRNA. The c-MYC downregulation resulted in the inhibition of cell proliferation and cell cycle arrest at G2/M phase, which was associated with decreased expression of cyclin B and cyclin-dependent kinase 1 (CDK1) and increased expression of CDK inhibitor p21 proteins. In addition, downregulation of c-MYC induced cell apoptosis characterized by DNA fragmentation and caspase-3 activation. Taken together, these results suggest that lentiviral vector-mediated siRNA for c-MYC may be a promising approach for targeting c-MYC in the treatment of Burkitt lymphoma.
Collapse
Affiliation(s)
- Aiqin Song
- Department of Pediatric Hematology, Affiliated Hospital of Qingdao University Medical College, 16 Jiangsu Road, Qingdao, 266001 Shandong, China.
| | | | | | | | | | | |
Collapse
|
73
|
Michel J, Cuchillo R. The impact of small molecule binding on the energy landscape of the intrinsically disordered protein C-myc. PLoS One 2012; 7:e41070. [PMID: 22815918 PMCID: PMC3397933 DOI: 10.1371/journal.pone.0041070] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 06/18/2012] [Indexed: 11/18/2022] Open
Abstract
Intrinsically disordered proteins are attractive therapeutic targets owing to their prevalence in several diseases. Yet their lack of well-defined structure renders ligand discovery a challenging task. An intriguing example is provided by the oncoprotein c-Myc, a transcription factor that is over expressed in a broad range of cancers. Transcriptional activity of c-Myc is dependent on heterodimerization with partner protein Max. This protein-protein interaction is disrupted by the small molecule 10058-F4 (1), that binds to monomeric and disordered c-Myc. To rationalize the mechanism of inhibition, structural ensembles for the segment of the c-Myc domain that binds to 1 were computed in the absence and presence of the ligand using classical force fields and explicit solvent metadynamics molecular simulations. The accuracy of the computed structural ensembles was assessed by comparison of predicted and measured NMR chemical shifts. The small molecule 1 was found to perturb the composition of the apo equilibrium ensemble and to bind weakly to multiple distinct c-Myc conformations. Comparison of the apo and holo equilibrium ensembles reveals that the c-Myc conformations binding 1 are already partially formed in the apo ensemble, suggesting that 1 binds to c-Myc through an extended conformational selection mechanism. The present results have important implications for rational ligand design efforts targeting intrinsically disordered proteins.
Collapse
Affiliation(s)
- Julien Michel
- EastCHEM School of Chemistry, University of Edinburgh, Edinburgh, United Kingdom.
| | | |
Collapse
|
74
|
Transcriptional regulators in hepatocarcinogenesis--key integrators of malignant transformation. J Hepatol 2012; 57:186-95. [PMID: 22446689 DOI: 10.1016/j.jhep.2011.11.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 11/28/2011] [Accepted: 11/30/2011] [Indexed: 12/26/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent human malignancies with poor prognosis and increasing incidence in the Western world. Only for a minority of HCC patients, surgical treatment options offer potential cure and therapeutic success of pharmacological approaches is limited. Highly specific approaches (e.g., kinase inhibitors) did not significantly improve the situation so far, possibly due to functional compensation, genetic heterogeneity of HCC, and development of resistance under selective pressure. In contrast, transcriptional regulators (especially transcription factors and co-factors) may integrate and process input signals of different (oncogenic) pathways and therefore represent cellular bottlenecks that regulate tumor cell biology. In this review, we want to summarize the current knowledge about central transcriptional regulators in human hepatocarcinogenesis and their potential as therapeutic target structures. Genomic and transcriptomic data of primary human HCC revealed that many of these factors showed up in subgroups of HCCs with a more aggressive phenotype, suggesting that aberrant activity of transcriptional regulators collect input information to promote tumor initiation and progression. Therefore, expression and dysfunction of transcription factors and co-factors may gain relevance for diagnostics and therapy of HCC.
Collapse
|
75
|
Zhang QL, Luo CL, Wu XH, Wang CY, Xu X, Zhang YY, Liu Q, Shen SL. HepaCAM induces G1 phase arrest and promotes c-Myc degradation in human renal cell carcinoma. J Cell Biochem 2012; 112:2910-9. [PMID: 21618595 DOI: 10.1002/jcb.23207] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hepatocyte cell adhesion molecule (hepaCAM) encodes a generally inactive phosphorylated glycoprotein which mediates cancer cell proliferation, migration, and differentiation. We have reported that hepaCAM is down-regulated in renal cell carcinoma (RCC) and takes responsibility of cell growth inhibition. However, the precise mechanisms of hepaCAM inhibits cell growth is still unknown. In this study, we demonstrated that re-expression of hepaCAM can cause an accumulation in G0/G1 phase in 786-0 cells. This reaction was accompanied by a substantial reduction of c-Myc expression through using an ectopic hepaCAM expression system. Furthermore, we found a comparable decrease in proliferation and G0/G1 accumulation of 786-0 and RC-2 cells after treatment with a small molecule c-Myc inhibitor, 10058-F4. This indicated that the down regulation of c-Myc was an essential process in controlling growth inhibitory actions of hepaCAM. Nevertheless, re-expression of hepaCAM results in apparent reduction of c-Myc protein with no corresponding reduction of c-Myc mRNA. This suggests that this reaction might take place at a post-transcriptional level rather than transcriptional one. Consistent with these findings, hepaCAM decreased c-Myc stability by increasing the proportion of c-Myc phosphorylation on T58 which can be abrogated by a proteasomal inhibitor (MG132). Thus, our research implies that the decrease in c-Myc protein expression, resulting from ectopic expression of hepaCAM, may contribute to the inhibition of proliferation in these cells.
Collapse
Affiliation(s)
- Qiao-Lin Zhang
- Department of Laboratory Diagnosis, Chongqing Medical University, Chongqing 400016, China
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Sanders JA, Schorl C, Patel A, Sedivy JM, Gruppuso PA. Postnatal liver growth and regeneration are independent of c-myc in a mouse model of conditional hepatic c-myc deletion. BMC PHYSIOLOGY 2012; 12:1. [PMID: 22397685 PMCID: PMC3353165 DOI: 10.1186/1472-6793-12-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 03/07/2012] [Indexed: 12/27/2022]
Abstract
BACKGROUND The transcription factor c-myc regulates genes involved in hepatocyte growth, proliferation, metabolism, and differentiation. It has also been assigned roles in liver development and regeneration. In previous studies, we made the unexpected observation that c-Myc protein levels were similar in proliferating fetal liver and quiescent adult liver with c-Myc displaying nucleolar localization in the latter. In order to investigate the functional role of c-Myc in adult liver, we have developed a hepatocyte-specific c-myc knockout mouse, c-mycfl/fl;Alb-Cre. RESULTS Liver weight to body weight ratios were similar in control and c-myc deficient mice. Liver architecture was unaffected. Conditional c-myc deletion did not result in compensatory induction of other myc family members or in c-Myc's binding partner Max. Floxed c-myc did have a negative effect on Alb-Cre expression at 4 weeks of age. To explore this relationship further, we used the Rosa26 reporter line to assay Cre activity in the c-myc floxed mice. No significant difference in Alb-Cre activity was found between control and c-mycfl/fl mice. c-myc deficient mice were studied in a nonproliferative model of liver growth, fasting for 48 hr followed by a 24 hr refeeding period. Fasting resulted in a decrease in liver mass and liver protein, both of which recovered upon 24 h of refeeding in the c-mycfl/fl;Alb-Cre animals. There was also no effect of reducing c-myc on recovery of liver mass following 2/3 partial hepatectomy. CONCLUSIONS c-Myc appears to be dispensable for normal liver growth during the postnatal period, restoration of liver mass following partial hepatectomy and recovery from fasting.
Collapse
Affiliation(s)
- Jennifer A Sanders
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI 02903, USA.
| | | | | | | | | |
Collapse
|
77
|
Chang CY, Kazmin D, Jasper JS, Kunder R, Zuercher WJ, McDonnell DP. The metabolic regulator ERRα, a downstream target of HER2/IGF-1R, as a therapeutic target in breast cancer. Cancer Cell 2011; 20:500-10. [PMID: 22014575 PMCID: PMC3199323 DOI: 10.1016/j.ccr.2011.08.023] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 01/27/2011] [Accepted: 08/26/2011] [Indexed: 01/28/2023]
Abstract
A genomic signature designed to assess the activity of the estrogen-related receptor alpha (ERRα) was used to profile more than 800 breast tumors, revealing a shorter disease-free survival in patients with tumors exhibiting elevated receptor activity. Importantly, this signature also predicted the ability of an ERRα antagonist, XCT790, to inhibit proliferation in cellular models of breast cancer. Using a chemical genomic approach, it was determined that activation of the Her2/IGF-1R signaling pathways and subsequent C-MYC stabilization upregulate the expression of peroxisome proliferator-activated receptor gamma coactivator-1 beta (PGC-1β), an obligate cofactor for ERRα activity. PGC-1β knockdown in breast cancer cells impaired ERRα signaling and reduced cell proliferation, implicating a functional role for PGC-1β/ERRα in the pathogenesis of breast cancers.
Collapse
Affiliation(s)
- Ching-yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dmitri Kazmin
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jeff S. Jasper
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Rebecca Kunder
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - William J. Zuercher
- Department of Chemical Biology, GlaxoSmithKline, Research Triangle Park, NC 27709, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
78
|
Porporato PE, Dhup S, Dadhich RK, Copetti T, Sonveaux P. Anticancer targets in the glycolytic metabolism of tumors: a comprehensive review. Front Pharmacol 2011; 2:49. [PMID: 21904528 PMCID: PMC3161244 DOI: 10.3389/fphar.2011.00049] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/05/2011] [Indexed: 12/21/2022] Open
Abstract
CANCER IS A METABOLIC DISEASE AND THE SOLUTION OF TWO METABOLIC EQUATIONS: to produce energy with limited resources and to fulfill the biosynthetic needs of proliferating cells. Both equations are solved when glycolysis is uncoupled from oxidative phosphorylation in the tricarboxylic acid cycle, a process known as the glycolytic switch. This review addresses in a comprehensive manner the main molecular events accounting for high-rate glycolysis in cancer. It starts from modulation of the Pasteur Effect allowing short-term adaptation to hypoxia, highlights the key role exerted by the hypoxia-inducible transcription factor HIF-1 in long-term adaptation to hypoxia, and summarizes the current knowledge concerning the necessary involvement of aerobic glycolysis (the Warburg effect) in cancer cell proliferation. Based on the many observations positioning glycolysis as a central player in malignancy, the most advanced anticancer treatments targeting tumor glycolysis are briefly reviewed.
Collapse
Affiliation(s)
- Paolo E Porporato
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research, University of Louvain Medical School Brussels, Belgium
| | | | | | | | | |
Collapse
|
79
|
Huang RS, Gamazon ER, Ziliak D, Wen Y, Im HK, Zhang W, Wing C, Duan S, Bleibel WK, Cox NJ, Dolan ME. Population differences in microRNA expression and biological implications. RNA Biol 2011; 8:692-701. [PMID: 21691150 DOI: 10.4161/rna.8.4.16029] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Population differences observed for complex traits may be attributed to the combined effect of socioeconomic, environmental, genetic and epigenetic factors. To better understand population differences in complex traits, genome-wide genetic and gene expression differences among ethnic populations have been studied. Here we set out to evaluate population differences in small non-coding RNAs through an evaluation of microRNA (miRNA) baseline expression in HapMap lymphoblastoid cell lines (LCLs) derived from 53 CEU (Utah residents with northern and western European ancestry) and 54 YRI (African from Ibadan, Nigeria). Using the Exiqon miRCURYTM LNA arrays, we found that 16% of all miRNAs evaluated in our study differ significantly between these 2 ethnic groups (pBonferroni corrected< 0.05). Furthermore, we explored the potential biological function of these observed differentially expressed miRNAs by comprehensively examining their effect on the transcriptome and their relationship with cellular sensitivity drug phenotypes. After multiple testing adjustment (false discovery rate (FDR)< 0.1), we found that 55% and 88% of the differentially expressed miRNAs were significantly and inversely correlated with an mRNA expression phenotype in the CEU and YRI samples, respectively. Interestingly, a substantial proportion (64%) of these miRNAs correlated with cellular sensitivity to chemotherapeutic agents (FDR< 0.05). Lastly, upon performing a genome-wide association study between SNPs and miRNA expression, we identified a large number of SNPs exhibiting different allele frequencies that affect the expression of these differentially expressed miRNAs, suggesting the role of genetic variants in mediating the observed population differences.
Collapse
Affiliation(s)
- R Stephanie Huang
- Section of Hematology-Oncology, University of Illinois at Chicago, Chicago, IL, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Clausen DM, Guo J, Parise RA, Beumer JH, Egorin MJ, Lazo JS, Prochownik EV, Eiseman JL. In vitro cytotoxicity and in vivo efficacy, pharmacokinetics, and metabolism of 10074-G5, a novel small-molecule inhibitor of c-Myc/Max dimerization. J Pharmacol Exp Ther 2010; 335:715-27. [PMID: 20801893 PMCID: PMC2993546 DOI: 10.1124/jpet.110.170555] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 08/24/2010] [Indexed: 01/29/2023] Open
Abstract
The c-Myc oncoprotein is overexpressed in many tumors and is essential for maintaining the proliferation of transformed cells. To function as a transcription factor, c-Myc must dimerize with Max via the basic helix-loop-helix leucine zipper protein (bHLH-ZIP) domains in each protein. The small molecule 7-nitro-N-(2-phenylphenyl)-2,1,3-benzoxadiazol-4-amine (10074-G5) binds to and distorts the bHLH-ZIP domain of c-Myc, thereby inhibiting c-Myc/Max heterodimer formation and inhibiting its transcriptional activity. We report in vitro cytotoxicity and in vivo efficacy, pharmacodynamics, pharmacokinetics, and metabolism of 10074-G5 in human xenograft-bearing mice. In vitro, 10074-G5 inhibited the growth of Daudi Burkitt's lymphoma cells and disrupted c-Myc/Max dimerization. 10074-G5 had no effect on the growth of Daudi xenografts in C.B-17 SCID mice that were treated with 20 mg/kg 10074-G5 intravenously for 5 consecutive days. Inhibition of c-Myc/Max dimerization in Daudi xenografts was not seen 2 or 24 h after treatment. Concentrations of 10074-G5 in various matrices were determined by high-performance liquid chromatography-UV, and metabolites of 10074-G5 were identified by liquid chromatography/tandem mass spectrometry. The plasma half-life of 10074-G5 in mice treated with 20 mg/kg i.v. was 37 min, and peak plasma concentration was 58 μM, which was 10-fold higher than peak tumor concentration. The lack of antitumor activity probably was caused by the rapid metabolism of 10074-G5 to inactive metabolites, resulting in tumor concentrations of 10074-G5 insufficient to inhibit c-Myc/Max dimerization. Our identification of 10074-G5 metabolites in mice will help design new, more metabolically stable small-molecule inhibitors of c-Myc.
Collapse
Affiliation(s)
- Dana M Clausen
- Molecular Therapeutics/Drug Discovery Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Kim J, Roh M, Abdulkadir SA. Pim1 promotes human prostate cancer cell tumorigenicity and c-MYC transcriptional activity. BMC Cancer 2010; 10:248. [PMID: 20515470 PMCID: PMC2886047 DOI: 10.1186/1471-2407-10-248] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 06/01/2010] [Indexed: 01/02/2023] Open
Abstract
Background The serine/threonine kinase PIM1 has been implicated as an oncogene in various human cancers including lymphomas, gastric, colorectal and prostate carcinomas. In mouse models, Pim1 is known to cooperate with c-Myc to promote tumorigenicity. However, there has been limited analysis of the tumorigenic potential of Pim1 overexpression in benign and malignant human prostate cancer cells in vivo. Methods We overexpressed Pim1 in three human prostate cell lines representing different disease stages including benign (RWPE1), androgen-dependent cancer (LNCaP) and androgen-independent cancer (DU145). We then analyzed in vitro and in vivo tumorigenicity as well as the effect of Pim1 overexpression on c-MYC transcriptional activity by reporter assays and gene expression profiling using an inducible MYC-ER system. To validate that Pim1 induces tumorigenicity and target gene expression by modulating c-MYC transcriptional activity, we inhibited c-MYC using a small molecule inhibitor (10058-F4) or RNA interference. Results Overexpression of Pim1 alone was not sufficient to convert the benign RWPE1 cell to malignancy although it enhanced their proliferation rates when grown as xenografts in vivo. However, Pim1 expression enhanced the in vitro and in vivo tumorigenic potentials of the human prostate cancer cell lines LNCaP and DU145. Reporter assays revealed increased c-MYC transcriptional activity in Pim1-expressing cells and mRNA expression profiling demonstrated that a large fraction of c-MYC target genes were also regulated by Pim1 expression. The c-MYC inhibitor 10058-F4 suppressed the tumorigenicity of Pim1-expressing prostate cancer cells. Interestingly, 10058-F4 treatment also led to a reduction of Pim1 protein but not mRNA. Knocking-down c-MYC using short hairpin RNA reversed the effects of Pim1 on Pim1/MYC target genes. Conclusion Our results suggest an in vivo role of Pim1 in promoting prostate tumorigenesis although it displayed distinct oncogenic activities depending on the disease stage of the cell line. Pim1 promotes tumorigenicity at least in part by enhancing c-MYC transcriptional activity. We also made the novel discovery that treatment of cells with the c-MYC inhibitor 10058-F4 leads to a reduction in Pim1 protein levels.
Collapse
Affiliation(s)
- Jongchan Kim
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | |
Collapse
|
82
|
Albihn A, Johnsen JI, Henriksson MA. MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res 2010; 107:163-224. [PMID: 20399964 DOI: 10.1016/s0065-230x(10)07006-5] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
MYC proteins (c-MYC, MYCN, and MYCL) regulate processes involved in many if not all aspects of cell fate. Therefore, it is not surprising that the MYC genes are deregulated in several human neoplasias as a result from genetic and epigenetic alterations. The near "omnipotency" together with the many levels of regulation makes MYC an attractive target for tumor intervention therapy. Here, we summarize some of the current understanding of MYC function and provide an overview of different cancer forms with MYC deregulation. We also describe available treatments and highlight novel approaches in the pursuit for MYC-targeting therapies. These efforts, at different stages of development, constitute a promising platform for novel, more specific treatments with fewer side effects. If successful a MYC-targeting therapy has the potential for tailored treatment of a large number of different tumors.
Collapse
Affiliation(s)
- Ami Albihn
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
83
|
Grewal S, Merchant T, Reymond R, McInerney M, Hodge C, Shearer P. Auditory late effects of childhood cancer therapy: a report from the Children's Oncology Group. Pediatrics 2010; 125:e938-50. [PMID: 20194279 PMCID: PMC3106205 DOI: 10.1542/peds.2009-1597] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Children treated for malignancies may be at risk for early- or delayed-onset hearing loss that can affect learning, communication, school performance, social interaction, and overall quality of life. Survivors at particular risk include those treated with platinum compounds (cisplatin and/or carboplatin) for neuroblastoma, hepatoblastoma, osteosarcoma, or germ-cell tumors and/or those treated with radiation that affects the ear at doses of >30 Gy for pediatric head and neck tumors. The aims of the Auditory/Hearing Late Effects Task Force of the Children's Oncology Group in this report were to (1) review ototoxicity resulting from childhood cancer therapy including platinum compounds (cisplatin and carboplatin) and radiation, (2) describe briefly cochlear pathophysiology and genetics of cisplatin-related hearing loss, (3) explain the impact of hearing loss resulting from chemotherapy and radiation, and (4) offer recommendations regarding evaluation and management of pediatric patients who are at risk for treatment-related hearing loss. A questionnaire is included as a tool to assist pediatricians in assessment.
Collapse
Affiliation(s)
- Satkiran Grewal
- Baystate Medical Center, Department of Pediatrics, Division of Pediatric Hematology/Oncology, Springfield, MA
| | - Thomas Merchant
- St. Jude Children’s Research Hospital, Department of Radiological Sciences, University of Tennessee College of Medicine, Memphis, TN
| | - Renee Reymond
- Ochsner Health Systems, Department of Pediatrics, New Orleans, LA
| | - Maryrose McInerney
- St John’s University, Department of Speech, Communication Sciences, & Theatre, Long Island AuD Consortium, Jamaica, NY
| | | | - Patricia Shearer
- University of Florida Shands Cancer Center, Gainesville, Florida
| |
Collapse
|
84
|
Lin CP, Liu CR, Lee CN, Chan TS, Liu HE. Targeting c-Myc as a novel approach for hepatocellular carcinoma. World J Hepatol 2010; 2:16-20. [PMID: 21160952 PMCID: PMC2999263 DOI: 10.4254/wjh.v2.i1.16] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2009] [Revised: 01/08/2010] [Accepted: 01/15/2010] [Indexed: 02/06/2023] Open
Abstract
Hepatocelluar carcinoma (HCC) is the most lethal cancer in the world. Most HCC over-express c-Myc, which plays a critical role in regulating cellular growth, differentiation and apoptosis in both normal and neoplastic cells. c-Myc is among the most frequently overexpressed genes in human cancers. Overexpression of c-Myc in hepatic cells leads to development of hepatocellular carcinoma. Here, we review the current progress in understanding physiologic function and regulation of c-Myc as well as its role in hepatic carcinogenesis and discuss the association of c-Myc activation in chronic hepatitis B infection and the upregulation of HIF-1/VEGF. We also explore the possibility of treating HCC by inhibiting c-Myc and examine the pros and cons of such an approach. Although this strategy is currently not available in clinics, with recent advances in better drug design, pharmacokinetics and pharmacogenetics, inhibition of c-Myc might become a novel therapy for HCC in the future.
Collapse
Affiliation(s)
- Che-Pin Lin
- Che-Pin Lin, Chien-Ru Liu, Department of Medicine, Taipei City Hospital, Ren-Ai Branch, Taipei 106, Taiwan
| | | | | | | | | |
Collapse
|
85
|
Takahashi K, Fukunaga K, Kobayashi A, Murata S, Oda T, Sasaki R, Ohkohchi N. A case of multiple hepatocellular carcinoma successfully treated by transcatheter arterial infusion chemotherapy and transcatheter arterial chemoembolization using IA-call®. KANZO 2010; 51:119-126. [DOI: 10.2957/kanzo.51.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
|
86
|
Xu J, Testa JR. DLX5 (distal-less homeobox 5) promotes tumor cell proliferation by transcriptionally regulating MYC. J Biol Chem 2009; 284:20593-601. [PMID: 19497851 DOI: 10.1074/jbc.m109.021477] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human DLX homeobox genes, which are related to Dll (Drosophila distal-less gene), encode transcription factors that are expressed primarily in embryonic development. Recently, DLX5 was reported to act as an oncogene in lymphomas and lung cancers, although the mechanism is not known. The identification of target genes of DLX5 can facilitate our understanding of oncogenic mechanisms driven by overexpression of DLX5. The MYC oncogene is aberrantly expressed in many human cancers and regulates transcription of numerous target genes involved in tumorigenesis. Here we demonstrate by luciferase assay that the MYC promoter is specifically activated by overexpression of DLX5 and that two DLX5 binding sites in the MYC promoter are important for transcriptional activation of MYC. We also show that DLX5 binds to the MYC promoter both in vitro and in vivo and that transfection of a DLX5 expression plasmid promotes the expression of MYC in a dose-dependent manner in mammalian cells. Furthermore, overexpression of DLX5 results in increased cell proliferation by up-regulating MYC. Knockdown of DLX5 in lung cancer cells overexpressing DLX5 resulted in decreased expression of MYC and reduced cell proliferation, which was rescued by overexpression of MYC. Because DLX5 has a restricted pattern of expression in adult tissues, it may serve as a potential therapeutic target for the treatment of cancers that overexpress DLX5.
Collapse
Affiliation(s)
- Jinfei Xu
- Cancer Signaling and Genetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | |
Collapse
|
87
|
Abstract
Medulloblastoma and neuroblastoma are malignant embryonal childhood tumours of the central and peripheral nervous systems, respectively, which often show poor clinical prognosis due to resistance to current chemotherapy. Both these tumours have deficient apoptotic machineries adopted from their respective progenitor cells. This review focuses on the specific background for tumour development, and highlights biological pathways that present potential targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- John Inge Johnsen
- Department of Woman and Child Health, Karolinska Institutet, Astrid Lindgren Children's Hospital, Childhood Cancer Research Unit, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|
88
|
Guo J, Parise RA, Joseph E, Egorin MJ, Lazo JS, Prochownik EV, Eiseman JL. Efficacy, pharmacokinetics, tisssue distribution, and metabolism of the Myc-Max disruptor, 10058-F4 [Z,E]-5-[4-ethylbenzylidine]-2-thioxothiazolidin-4-one, in mice. Cancer Chemother Pharmacol 2009; 63:615-25. [PMID: 18509642 PMCID: PMC2752825 DOI: 10.1007/s00280-008-0774-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Accepted: 05/09/2008] [Indexed: 12/28/2022]
Abstract
OBJECTIVES c-Myc is commonly activated in many human tumors and is functionally important in cellular proliferation, differentiation, apoptosis and cell cycle progression. The activity of c-Myc requires noncovalent interaction with its client protein Max. In vitro studies indicate the thioxothiazolidinone, 10058-F4, inhibits c-Myc/Max dimerization. In this study, we report the efficacy, pharmacokinetics and metabolism of this novel protein-protein disruptor in mice. METHODS SCID mice bearing DU145 or PC-3 human prostate cancer xenografts were treated with either 20 or 30 mg/kg 10058-F4 on a qdx5 schedule for 2 weeks for efficacy studies. For pharmacokinetics and metabolism studies, mice bearing PC-3 or DU145 xenografts were treated with 20 mg/kg of 10058-F4 i.v. Plasma and tissues were collected 5-1440 min after dosing. The concentration of 10058-F4 in plasma and tissues was determined by HPLC, and metabolites were characterized by LC-MS/MS. RESULTS Following a single iv dose, peak plasma 10058-F4 concentrations of approximately 300 muM were seen at 5 min and declined to below the detection limit at 360 min. Plasma concentration versus time data were best approximated by a two-compartment, open, linear model. The highest tissue concentrations of 10058-F4 were found in fat, lung, liver, and kidney. Peak tumor concentrations of 10058-F4 were at least tenfold lower than peak plasma concentrations. Eight metabolites of 10058-F4 were identified in plasma, liver, and kidney. The terminal half-life of 10058-F4 was approximately 1 h, and the volume of distribution was >200 ml/kg. No significant inhibition of tumor growth was seen after i.v. treatment of mice with either 20 or 30 mg/kg 10058-F4. CONCLUSION The lack of significant antitumor activity of 10058-F4 in tumor-bearing mice may have resulted from its rapid metabolism and low concentration in tumors.
Collapse
Affiliation(s)
- Jianxia Guo
- Hillman Cancer Center, The University of Pittsburgh Cancer Institute, Room G27b. 5117 Centre Ave., Pittsburgh, PA 15213, USA, e-mail: ;
| | - Robert A. Parise
- Hillman Cancer Center, The University of Pittsburgh Cancer Institute, Room G27b. 5117 Centre Ave., Pittsburgh, PA 15213, USA, e-mail: ;
| | - Erin Joseph
- Hillman Cancer Center, The University of Pittsburgh Cancer Institute, Room G27b. 5117 Centre Ave., Pittsburgh, PA 15213, USA, e-mail: ;
| | - Merrill J. Egorin
- Hillman Cancer Center, The University of Pittsburgh Cancer Institute, Room G27b. 5117 Centre Ave., Pittsburgh, PA 15213, USA, e-mail: ;
| | - John S. Lazo
- Department of Pharmacology, The University of Pittsburgh Cancer Institute, Room G27b. 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | | | - Julie L. Eiseman
- Hillman Cancer Center, The University of Pittsburgh Cancer Institute, Room G27b. 5117 Centre Ave., Pittsburgh, PA 15213, USA, e-mail: ;
| |
Collapse
|
89
|
Nakai T, Mochida J, Sakai D. Synergistic role of c-Myc and ERK1/2 in the mitogenic response to TGF beta-1 in cultured rat nucleus pulposus cells. Arthritis Res Ther 2008; 10:R140. [PMID: 19061498 PMCID: PMC2656245 DOI: 10.1186/ar2567] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 11/29/2008] [Accepted: 12/05/2008] [Indexed: 12/24/2022] Open
Abstract
Introduction Although transforming growth factor β1 (TGFβ1) is known to be a potent inhibitor of proliferation in most cell types, it accelerates proliferation in certain mesenchymal cells, such as articular chondrocytes and nucleus pulposus cells. The low ability for self-renewal of nucleus pulposus cells is one obstacle in developing new therapeutic options for intervertebral disc diseases, and utilizing cytokines is one of the strategies to regulate nucleus pulposus cell proliferation. However, the precise cell cycle progression and molecular mechanisms by which TGFβ1 stimulates cell growth remain unclear. The aim of this study was to elucidate a mechanism that enables cell proliferation with TGFβ1 stimulation. Methods We tested cultured rat nucleus pulposus cells for proliferation and cell cycle distribution under exogenous TGFβ1 stimulation with and without putative pharmaceutical inhibitors. To understand the molecular mechanism, we evaluated the expression levels of key regulatory G1 phase proteins, c-Myc and the cyclin-dependent kinase inhibitors. Results We found that TGFβ1 promoted proliferation and cell cycle progression while reducing expression of the cyclin-dependent kinase inhibitors p21 and p27, which are downregulators of the cell cycle. Robust c-Myc expression for 2 h and immediate phosphorylation of extra cellular signal regulated kinase (ERK1/2) were detected in cultures when TGFβ1 was added. However, pretreatment with 10058-F4 (an inhibitor of c-Myc transcriptional activity) or PD98059 (an inhibitor of ERK1/2) suppressed c-Myc expression and ERK1/2 phosphorylation, and inhibited cell cycle promotion by TGFβ1. Conclusions Our experimental results indicate that TGFβ1 promotes cell proliferation and cell cycle progression in rat nucleus pulposus cells and that c-Myc and phosphorylated ERK1/2 play important roles in this mechanism. While the difference between rat and human disc tissues requires future studies using different species, investigation of distinct response in the rat model provides fundamental information to elucidate a specific regulatory pathway of TGFβ1.
Collapse
Affiliation(s)
- Tomoko Nakai
- Division of Organogenesis, Research Center for Regenerative Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan.
| | | | | |
Collapse
|
90
|
Liu X, Chen LB, Ye J, Jiang J, Xu JY, Qian W, 钱 伟. Effects of shRNA-induced SMYD3 silence on c-Myc expression and apoptosis of HepG2 cells. Shijie Huaren Xiaohua Zazhi 2008; 16:1373-1377. [DOI: 10.11569/wcjd.v16.i13.1373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the c-Myc gene expression and apoptosis of HepG2 cells after SET and MYND-domain containing 3 (SMYD3) silence induced by short hairpin RNA.
METHODS: Three short hairpin RNA interference plasmids targeting SMYD3 were prepared as 3 groups: Pgenesil-1-s1 (with interfering effect), Pgenesil-1-s2 (with interfering effect), and Pgenesil-1-hk (negative control plasmid, without interfering effect). Meanwhile, an empty control group was also designed. Transfection was performed using the Lipofectmine2000 liposome. Reverse transcription-polymerase chain reaction (RT-PCR) was employed to detect the expression of SMYD3 and c-Myc gene 24, 48 and 72 h after transfection. Flow cytometry (FCM) was used to detect cell apoptosis in each group.
RESULTS: SMYD3 and c-Myc gene were strongly expressed in HepG2 cells. The expression of SMYD3 gene was significantly inhibited after 24-, 48- and 72-h transfection (F = 67.46, P < 0.01; F = 176.79, P < 0.01; F = 175.28, P < 0.01). At the same time, c-Myc gene mRNA expression was down-regulated in the Pgenesil-1 transfected group as compared with that in the Pgenesil-1-hk group (F = 11.58, P = 0.009; F = 126.41, P < 0.01; F = 261.25, P < 0.01). Moreover, the early apoptosis rate was significantly higher in the Pgenesil-1-s1 or Pgenesil-1-s2 group than that in the Pgenesil-1-hk group (LSD-t = -13.58, -12.62; both P < 0.01) and the empty control group (LSD-t = -18.62, -17.67; P < 0.01).
CONCLUSION: Short hairpin RNA interference targeting SMYD3 may inhibit the expression of c-Myc gene in HepG2 cells, thus promoting the apoptosis of HepG2 cells.
Collapse
|
91
|
A dyskerin motif reactivates telomerase activity in X-linked dyskeratosis congenita and in telomerase-deficient human cells. Blood 2007; 111:2606-14. [PMID: 18057229 DOI: 10.1182/blood-2007-04-083261] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Dyskerin gene is mutated in patients with X-linked dyskeratosis congenita (X-DC), which results in greatly reduced levels of telomerase activity. A genetic suppressor element (GSE) termed GSE24-2 has been isolated in a screening for cisplatin resistance. GSE24-2-expressing cells presented impaired telomerase inhibition following in vitro exposure to chemotherapies, such as cisplatin, or telomerase inhibitors. The promoter of the telomerase component hTERT was constitutively activated in GSE24-2 cells in a c-myc expression-dependent manner. Deletion analyses and mutagenesis of the human c-myc promoter demonstrated that the target sequence for activation was the nuclease hypersensitive element-III (NHEIII) site located upstream to the P1 region of the promoter. Further, expression of GSE24-2 in cell lines derived from patients with X-DC and in VA13 cells induced increased hTERT RNA and hTR levels and recovery of telomerase activity. Finally, expression of GSE24-2 was able to rescue X-DC fibroblasts from premature senescence. These data demonstrate that this domain of dyskerin plays an important role in telomerase maintenance following cell insults such as cisplatin treatment, and in telomerase-defective cells in patients with X-DC. The expression of this dyskerin fragment has a dominant function in X-DC cells and could provide the basis for a therapeutic approach to this disease.
Collapse
|
92
|
Pastorino F, Mumbengegwi DR, Ribatti D, Ponzoni M, Allen TM. Increase of therapeutic effects by treating melanoma with targeted combinations of c-myc antisense and doxorubicin. J Control Release 2007; 126:85-94. [PMID: 18166243 DOI: 10.1016/j.jconrel.2007.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Accepted: 11/13/2007] [Indexed: 11/26/2022]
Abstract
Patients with advanced or metastatic melanoma have a very poor prognosis, due to the resistance of melanoma cells to conventional chemotherapy. We previously reported that coated cationic liposomes targeted with a monoclonal antibody against the disialoganglioside GD(2) and containing c-myc antisense oligodeoxynucleotides (alpha GD(2)-CCL[c-myc-as]) induced partial tumor growth arrest in melanoma xenografts. Here we addressed the role of c-myc-asODN treatment in the susceptibility to doxorubicin (DXR) in human melanoma cells. Cytotoxicity studies revealed that growth of melanoma cells was inhibited to a greater extent by alpha GD(2)-CCL[c-myc-as] than by the corresponding non-targeted formulations or by free c-myc-as. Targeted c-myc-as sensitized cells to DXR, reducing the IC(50) by approximately 10-fold. Scrambled ODNs had no effect on the IC(50) of DXR. Compared to either treatment alone, combination of targeted c-myc-as and DXR resulted in earlier apoptosis and in cell death after 2 days of treatment. In vivo experiments revealed that liposomal formulations of c-myc-as and DXR, both targeted via GD(2), led to the most pronounced delay in tumor growth when administered in a sequential manner. As a result, their combination translates into a statistically significant suppression of blood vessel density and an enhanced apoptosis, compared to all treatments given separately. Our data indicate the increasing cell sensitivity to DXR by c-myc-asODNs as a promising basis for developing novel anti-tumor strategy against advanced melanoma.
Collapse
Affiliation(s)
- Fabio Pastorino
- Differentiation Therapy Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, 16148-Genoa, Italy.
| | | | | | | | | |
Collapse
|
93
|
Abstract
Patients with an elevated serum total prostate-specific antigen value or abnormal digital rectal examination results are at risk of having prostate cancer and should undergo prostate needle biopsies. However, approximately 60% of them will have a negative prostate biopsy result. Therefore, further biopsies are recommended for young patients at risk of prostate cancer with a positive rate of 20-40%. Biomarkers are required in order to avoid unnecessary biopsies. The PCA3 gene product is specifically overexpressed in prostate tumor cells, and modern molecular biology techniques allow us to use a specific test for this gene in order to select patients who have a high risk of having prostate cancer. Literature reviews of the gene product, as well as the first clinical results of the Progensa PCA3 test, are presented.
Collapse
Affiliation(s)
- Alexandre de la Taille
- INSERM U841Eq07, Assistance Publique des Hôpitaux de Paris, Henri Mondor Hospital, Department of Urology, CHU Mondor, 51 avenue du Maréchal de Lattre de Tassigny, 94000, Créteil, France.
| |
Collapse
|
94
|
Huang RS, Duan S, Shukla SJ, Kistner EO, Clark TA, Chen TX, Schweitzer AC, Blume JE, Dolan ME. Identification of genetic variants contributing to cisplatin-induced cytotoxicity by use of a genomewide approach. Am J Hum Genet 2007; 81:427-37. [PMID: 17701890 PMCID: PMC1950832 DOI: 10.1086/519850] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Accepted: 05/15/2007] [Indexed: 01/17/2023] Open
Abstract
Cisplatin, a platinating agent commonly used to treat several cancers, is associated with nephrotoxicity, neurotoxicity, and ototoxicity, which has hindered its utility. To gain a better understanding of the genetic variants associated with cisplatin-induced toxicity, we present a stepwise approach integrating genotypes, gene expression, and sensitivity of HapMap cell lines to cisplatin. Cell lines derived from 30 trios of European descent (CEU) and 30 trios of African descent (YRI) were used to develop a preclinical model to identify genetic variants and gene expression that contribute to cisplatin-induced cytotoxicity in two different populations. Cytotoxicity was determined as cell-growth inhibition at increasing concentrations of cisplatin for 48 h. Gene expression in 176 HapMap cell lines (87 CEU and 89 YRI) was determined using the Affymetrix GeneChip Human Exon 1.0 ST Array. We identified six, two, and nine representative SNPs that contribute to cisplatin-induced cytotoxicity through their effects on 8, 2, and 16 gene expressions in the combined, Centre d'Etude du Polymorphisme Humain (CEPH), and Yoruban populations, respectively. These genetic variants contribute to 27%, 29%, and 45% of the overall variation in cell sensitivity to cisplatin in the combined, CEPH, and Yoruban populations, respectively. Our whole-genome approach can be used to elucidate the expression of quantitative trait loci contributing to a wide range of cellular phenotypes.
Collapse
Affiliation(s)
- R Stephanie Huang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
De Cian A, Lacroix L, Douarre C, Temime-Smaali N, Trentesaux C, Riou JF, Mergny JL. Targeting telomeres and telomerase. Biochimie 2007; 90:131-55. [PMID: 17822826 DOI: 10.1016/j.biochi.2007.07.011] [Citation(s) in RCA: 484] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Accepted: 07/16/2007] [Indexed: 01/06/2023]
Abstract
Telomeres and telomerase represent, at least in theory, an extremely attractive target for cancer therapy. The objective of this review is to present the latest view on the mechanism(s) of action of telomerase inhibitors, with an emphasis on a specific class of telomere ligands called G-quadruplex ligands, and to discuss their potential use in oncology.
Collapse
Affiliation(s)
- Anne De Cian
- INSERM, U565, Acides nucléiques: dynamique, ciblage et fonctions biologiques, 43 rue Cuvier, CP26, Paris Cedex 05, F-75231, France
| | | | | | | | | | | | | |
Collapse
|