201
|
Wang CA, Drasin D, Pham C, Jedlicka P, Zaberezhnyy V, Guney M, Li H, Nemenoff R, Costello JC, Tan AC, Ford HL. Homeoprotein Six2 promotes breast cancer metastasis via transcriptional and epigenetic control of E-cadherin expression. Cancer Res 2014; 74:7357-70. [PMID: 25348955 DOI: 10.1158/0008-5472.can-14-0666] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Misexpression of developmental transcription factors occurs often in human cancers, where embryonic programs may be reinstated in a context that promotes or sustains malignant development. In this study, we report the involvement of the kidney development transcription factor Six2 in the metastatic progression of human breast cancer. We found that Six2 promoted breast cancer metastasis by a novel mechanism involving both transcriptional and epigenetic regulation of E-cadherin. Downregulation of E-cadherin by Six2 was necessary for its ability to increase soft agar growth and in vivo metastasis in an immunocompetent mouse model of breast cancer. Mechanistic investigations showed that Six2 represses E-cadherin expression by upregulating Zeb2, in part, through a microRNA-mediated mechanism and by stimulating promoter methylation of the E-cadherin gene (Cdh1). Clinically, SIX2 expression correlated inversely with CDH1 expression in human breast cancer specimens, corroborating the disease relevance of their interaction. Our findings establish Six2 as a regulator of metastasis in human breast cancers and demonstrate an epigenetic function for SIX family transcription factors in metastatic progression through the regulation of E-cadherin.
Collapse
Affiliation(s)
- Chu-An Wang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| | - David Drasin
- Program in Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Catherine Pham
- Program in Cancer Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Paul Jedlicka
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Vadym Zaberezhnyy
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Michelle Guney
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| | - Howard Li
- Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Raphael Nemenoff
- Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - James C Costello
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| | - Aik-Choon Tan
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Heide L Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado. Program in Molecular Biology, University of Colorado School of Medicine, Aurora, Colorado. Program in Cancer Biology, University of Colorado School of Medicine, Aurora, Colorado. Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
202
|
Yu MO, Song NH, Park KJ, Park DH, Kim SH, Chae YS, Chung YG, Chi SG, Kang SH. Romo1 is associated with ROS production and cellular growth in human gliomas. J Neurooncol 2014; 121:73-81. [PMID: 25193023 DOI: 10.1007/s11060-014-1608-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 08/27/2014] [Indexed: 12/11/2022]
Abstract
Romo1 is a mitochondrial protein whose elevated expression is commonly observed in various types of human cancers. However, the expression status of Romo1 and its implication in the pathogenesis of human glioblastoma (GBM) remain largely undefined. To understand the role of Romo1 in the progression of GBM, we explored its expression in a series of GBM tissues and cell lines and determined its effect on ROS production, cell proliferation, and tumor growth. Romo1 was frequently overexpressed at the mRNA level in both primary tumors and cell lines and its elevation was more commonly observed in high grade tumors versus low grade tumors. Romo1 expression was associated with ROS production and its knockdown led to a marked reduction of in vitro cellular growth and anchorage-independent growth of GBM. Consistently, Romo1 depletion induced a G2/M arrest of the cell cycle that was accompanied with accumulation of phospho-cdc2. Furthermore, a mouse xenograft assay revealed that Romo1 depletion significantly decreased tumor formation and growth. Therefore, our data demonstrate that Romo1 upregulation is a common event in human GBMs and contributes to the malignant tumor progression, suggesting that Romo1 could be a new therapeutic target for human GBM.
Collapse
Affiliation(s)
- Mi Ok Yu
- Department of Neurosurgery College of Medicine, Korea University, #126, 5-ga, Anam-Dong, Seongbuk-Gu, Seoul, 136-705, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Expression of astrocyte elevated gene-1 (AEG-1) in human meningiomas and its roles in cell proliferation and survival. J Neurooncol 2014; 121:31-9. [DOI: 10.1007/s11060-014-1603-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/23/2014] [Indexed: 12/16/2022]
|
204
|
Soares AS, Costa VM, Diniz C, Fresco P. The combination of Cl-IB-MECA with paclitaxel: a new anti-metastatic therapeutic strategy for melanoma. Cancer Chemother Pharmacol 2014; 74:847-60. [PMID: 25119183 DOI: 10.1007/s00280-014-2557-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/26/2014] [Indexed: 12/18/2022]
Abstract
PURPOSE Metastatic melanoma is considered one of the most aggressive malignant tumours, representing the deadliest form of skin cancer. Melanoma progression is associated with the abrogation of normal controls that limit cell proliferation, migration, and invasion, eventually leading to metastasis. Based on the variety of cellular mechanisms involved in metastatic progression, we aimed to evaluate the effect of inosine (50 μM) and of the combination of Cl-IB-MECA (10 μM) with paclitaxel (10 ng/mL) on several stages of melanoma progression. METHODS Proliferation, migration, adhesion, invasion, and colony formation assays were performed on human C32 and A375 metastatic melanoma cells. Levels of ERK1/2 were also determined using an ELISA kit. Moreover, mouse aortic rings were treated with vascular endothelial growth factor in order to assess the microvessel sprouting (an indicator of angiogenesis) in the presence of the referred compounds. RESULTS We demonstrate that inosine induced, through A3 adenosine receptor activation, proliferation, migration, adhesion, and invasion on C32 and A375 melanoma cells, although with dissimilar importance on the two melanoma cell lines. Inosine also increased colony formation on A375 cells. Levels of ERK1/2 were increased after inosine exposure and that increase was dependent on A3 adenosine receptor activation in both cell lines. Moreover, microvessel sprouting stimulated by inosine was decreased by the combination of Cl-IB-MECA with paclitaxel. CONCLUSIONS Cl-IB-MECA combined with paclitaxel was able to impair almost all of the referred metastatic related mechanisms induced by inosine, making this approach a valuable tool for combinatory therapy against metastatic melanoma.
Collapse
Affiliation(s)
- Ana S Soares
- REQUIMTE/Laboratório de Farmacologia, Departamento de Ciências do Medicamento, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | | | | | | |
Collapse
|
205
|
Lien IC, Horng LY, Hsu PL, Wu CL, Sung HC, Wu RT. Internal ribosome entry site of bFGF is the target of thalidomide for IMiDs development in multiple myeloma. Genes Cancer 2014; 5:127-41. [PMID: 25053990 PMCID: PMC4091528 DOI: 10.18632/genesandcancer.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 05/20/2014] [Indexed: 01/20/2023] Open
Abstract
Although new analogues of immunomodulatory drugs (IMiDs) are being developed for MM, the molecular mechanism of these drugs remains unclear. In the current study, we used MM cell lines as a model to investigate the molecular mechanism of thalidomide and to compare its potency with IMiDs such as pomalidomide. We determined that thalidomide did not inhibit cell proliferation of RPMI8226 and U266 MM cells, whereas pomalidomide showed a significant inhibitory effect on these two MM cell lines. Interestingly, we further demonstrated that although thalidomide down-regulated bFGF translation through the inhibition of IRES even at 0.1 μg/ml, pomalidomide did not have a similar affect bFGF levels. A colony formation assay demonstrated that thalidomide and the bFGF knock-down clones caused a significant reduction in the clonogenic ability of MM cells, and treatment with exogenous bFGF can recover the clonogenic ability of thalidomide-treated cells and knock-down clones, but not that of pomalidomide-treated cells. This implies that thalidomide, but not pomalidomide, targets the IRES of FGF-2. In conclusion, our results highlight a non-cytotoxic anticancer drug target for thalidomide, the IRES of bFGF, and provide the mechanistic rationale for developing IMiDs as anti-cancer therapeutics in MM patients, with improved potency and fewer side effects.
Collapse
Affiliation(s)
- I-Chia Lien
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, ROC (Taiwan)
| | - Lin-Yea Horng
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, ROC (Taiwan) ; Research Centre for Drug Discovery, National Yang-Ming University, Taipei, ROC (Taiwan)
| | - Pei-Lun Hsu
- Research Centre for Drug Discovery, National Yang-Ming University, Taipei, ROC (Taiwan)
| | - Chia-Ling Wu
- Research Centre for Drug Discovery, National Yang-Ming University, Taipei, ROC (Taiwan)
| | - Hui-Ching Sung
- Research Centre for Drug Discovery, National Yang-Ming University, Taipei, ROC (Taiwan)
| | - Rong-Tsun Wu
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, ROC (Taiwan) ; Research Centre for Drug Discovery, National Yang-Ming University, Taipei, ROC (Taiwan)
| |
Collapse
|
206
|
RAGE overexpression confers a metastatic phenotype to the WM115 human primary melanoma cell line. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1017-27. [DOI: 10.1016/j.bbadis.2014.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 02/16/2014] [Accepted: 02/26/2014] [Indexed: 12/19/2022]
|
207
|
Lawrenson K, Lee N, Torres HAM, Lee JM, Brueggmann D, Rao PN, Noushmehr H, Gayther SA. Src as a novel therapeutic target for endometriosis. Gynecol Oncol 2014; 135:100-7. [PMID: 24972189 DOI: 10.1016/j.ygyno.2014.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/13/2014] [Accepted: 06/16/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Endometriosis is a common condition that is associated with an increased risk of developing ovarian carcinoma. Improved in vitro models of this disease are needed to better understand how endometriosis, a benign disease, can undergo neoplastic transformation, and for the development of novel treatment strategies to prevent this progression. METHODS We describe the generation and in vitro characterization of novel TERT immortalized ovarian endometriosis epithelial cell lines (EEC16-TERT). RESULTS Expression of TERT alone was sufficient to immortalize endometriosis epithelial cells. TERT immortalization induces an epithelial-to-mesenchymal transition and perturbation in the expression of genes involved in the development of ovarian cancer. EEC16-TERT was non-tumorigenic when xenografted into immunocompromised mice but grew in anchorage-independent growth assays in an epidermal growth factor and hydrocortisone dependent manner. Colony formation in agar was abolished by inhibition of Src, and the Src pathway was found to be activated in human endometriosis lesions. CONCLUSIONS This new in vitro model system mimics endometriosis and the early stages of neoplastic transformation in the development of endometriosis associated ovarian cancer. We demonstrate the potential clinical relevance of this model by identifying Src activation as a novel pathway in endometriosis that could be targeted therapeutically, perhaps as a novel strategy to manage endometriosis clinically, or to prevent the development of endometriosis-associated ovarian cancer.
Collapse
Affiliation(s)
- Kate Lawrenson
- Department of Preventive Medicine, University of Southern California/Keck School of Medicine, 1450 Biggy Street, Los Angeles, CA 90033, USA.
| | - Nathan Lee
- Department of Preventive Medicine, University of Southern California/Keck School of Medicine, 1450 Biggy Street, Los Angeles, CA 90033, USA.
| | - Hugo A M Torres
- Department of Genetics, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Núcleo de Apoio à Pesquisa (NAP) denominado Centro de Biologia Sistêmica Integrada (CISBi), São Paulo, Brazil
| | - Janet M Lee
- Department of Preventive Medicine, University of Southern California/Keck School of Medicine, 1450 Biggy Street, Los Angeles, CA 90033, USA.
| | - Doerthe Brueggmann
- Department of Obstetrics and Gynecology, University of Southern California/Keck School of Medicine, 1450 Biggy Street, Los Angeles, CA 90033, USA.
| | - P Nagesh Rao
- Pathology and Lab Medicine, David Geffen University of California Los Angeles, 22-26 Rehab Cntr, 1000 Veteran Ave, Los Angeles, CA 90024, USA.
| | - Houtan Noushmehr
- Department of Genetics, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Núcleo de Apoio à Pesquisa (NAP) denominado Centro de Biologia Sistêmica Integrada (CISBi), São Paulo, Brazil.
| | - Simon A Gayther
- Department of Preventive Medicine, University of Southern California/Keck School of Medicine, 1450 Biggy Street, Los Angeles, CA 90033, USA.
| |
Collapse
|
208
|
Peng HS, Liao MB, Zhang MY, Xie Y, Xu L, Zhang YJ, Zheng XFS, Wang HY, Chen YF. Synergistic inhibitory effect of hyperbaric oxygen combined with sorafenib on hepatoma cells. PLoS One 2014; 9:e100814. [PMID: 24956259 PMCID: PMC4067386 DOI: 10.1371/journal.pone.0100814] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 05/29/2014] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Hypoxia is a common phenomenon in solid tumors, associated with chemotherapy and radiotherapy resistance, recurrence and metastasis. Hyperbaric oxygen (HBO) therapy can increase tissue oxygen pressure and content to prevent the resistance, recurrence and metastasis of cancer. Presently, Sorafenib is a first-line drug, targeted for hepatocellular carcinoma (HCC) but effective in only a small portion of patients and can induce hypoxia. The purpose of this study is to investigate the effect of HBO in combination with sorafenib on hepatoma cells. METHODS Hepatoma cell lines (BEL-7402 and SK-Hep1) were treated with HBO at 2 atmosphere absolute pressure for 80 min per day or combined with sorafenib or cisplatin. At different time points, cells were tested for cell growth, colony formation, apoptosis, cell cycle and migration. Finally, miRNA from the hepatoma cells was detected by microRNA array and validated by qRT-PCR. RESULTS Although HBO, sorafenib or cisplatin alone could inhibit growth of hepatoma cells, HBO combined with sorafenib or cisplatin resulted in much greater synergistic growth inhibition (cell proliferation and colony formation) in hepatoma cells. Similarly, the synergistic effect of HBO and sorafenib on induction of apoptosis was also observed in hepatoma cells. HBO induced G1 arrest in SK-Hep1 not in BEL-7402 cells, but enhanced cell cycle arrest induced by sorafenib in BEL-7402 treated cells. However, HBO had no obvious effect on the migration of hepatoma cells, and microRNA array analysis showed that hepatoma cells with HBO treatment had significantly different microRNA expression profiles from those with blank control. CONCLUSIONS We show for the first time that HBO combined with sorafenib results in synergistic growth inhibition and apoptosis in hepatoma cells, suggesting a potential application of HBO combined with sorafenib in HCC patients. Additionally, we also show that HBO significantly altered microRNA expression in hepatoma cells.
Collapse
Affiliation(s)
- Hai-Shan Peng
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ming-Bin Liao
- Hyperbaric Oxygen Therapy Center, Affiliated Guangzhou First People's Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yin Xie
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Li Xu
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yao-Jun Zhang
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - X. F. Steven Zheng
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- * E-mail: (YFC); (HYW)
| | - Yi-Fei Chen
- Hyperbaric Oxygen Therapy Center, Affiliated Guangzhou First People's Hospital of Guangzhou Medical University, Guangzhou, China
- * E-mail: (YFC); (HYW)
| |
Collapse
|
209
|
ICAM-2 confers a non-metastatic phenotype in neuroblastoma cells by interaction with α-actinin. Oncogene 2014; 34:1553-62. [PMID: 24704826 DOI: 10.1038/onc.2014.87] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 01/29/2014] [Accepted: 02/24/2014] [Indexed: 12/16/2022]
Abstract
Progressive metastatic disease is a major cause of mortality for patients diagnosed with multiple types of solid tumors. One of the long-term goals of our laboratory is to identify molecular interactions that regulate metastasis, as a basis for developing agents that inhibit this process. Toward this goal, we recently demonstrated that intercellular adhesion molecule-2 (ICAM-2) converted neuroblastoma (NB) cells from a metastatic to a non-metastatic phenotype, a previously unknown function for ICAM-2. Interestingly, ICAM-2 suppressed metastatic but not tumorigenic potential in preclinical models, supporting a novel mechanism of regulating metastasis. We hypothesized that the effects of ICAM-2 on NB cell phenotype depend on the interaction of ICAM-2 with the cytoskeletal linker protein α-actinin. The goal of the study presented here was to evaluate the impact of α-actinin binding to ICAM-2 on the phenotype of NB tumor cells. We used in silico approaches to examine the likelihood that the cytoplasmic domain of ICAM-2 binds directly to α-actinin. We then expressed variants of ICAM-2 with mutated α-actinin-binding domains, and compared the impact of ICAM-2 and each variant on NB cell adhesion, migration, anchorage-independent growth, co-precipitation with α-actinin and production of localized and disseminated tumors in vivo. The in vitro and in vivo characteristics of cells expressing ICAM-2 variants with modified α-actinin-binding domains differed from cells expressing ICAM-2 wild type (WT) and also from cells that expressed no detectable ICAM-2. Like the WT protein, ICAM-2 variants inhibited cell adhesion, migration and colony growth in vitro. However, unlike the WT protein, ICAM-2 variants did not completely suppress development of disseminated NB tumors in vivo. The data suggest the presence of α-actinin-dependent and α-actinin-independent mechanisms, and indicate that the interaction of ICAM-2 with α-actinin is critical to conferring an ICAM-2-mediated non-metastatic phenotype in NB cells.
Collapse
|
210
|
Polvani S, Tarocchi M, Tempesti S, Mello T, Ceni E, Buccoliero F, D'Amico M, Boddi V, Farsi M, Nesi S, Nesi G, Milani S, Galli A. COUP-TFII in pancreatic adenocarcinoma: clinical implication for patient survival and tumor progression. Int J Cancer 2014; 134:1648-58. [PMID: 24122412 DOI: 10.1002/ijc.28502] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 08/10/2013] [Accepted: 09/06/2013] [Indexed: 12/16/2023]
Abstract
Despite the accumulating knowledge of alterations in pancreatic cancer molecular pathways, no substantial improvements in the clinical prognosis have been made and this malignancy continues to be a leading cause of cancer death in the Western World. The orphan nuclear receptor COUP-TFII is a regulator of a wide range of biological processes and it may exert a pro-oncogenic role in cancer cells; interestingly, indirect evidences suggest that the receptor could be involved in pancreatic cancer. The aim of this study was to evaluate the expression of COUP-TFII in human pancreatic tumors and to unveil its role in the regulation of pancreatic tumor growth. We evaluated COUP-TFII expression by immunohistochemistry on primary samples. We analyzed the effect of the nuclear receptor silencing in human pancreatic cancer cells by means of shRNA expressing cell lines. We finally confirmed the in vitro results by in vivo experiments on nude mice. COUP-TFII is expressed in 69% of tested primary samples and correlates with the N1 and M1 status and clinical stage; Kaplan-Meier and Cox regression analysis show that it may be an independent prognostic factor of worst outcome. In vitro silencing of COUP-TFII reduces the cell growth and invasiveness and it strongly inhibits angiogenesis, an effect mediated by the regulation of VEGF-C. In nude mice, COUP-TFII silencing reduces tumor growth by 40%. Our results suggest that COUP-TFII might be an important regulator of the behavior of pancreatic adenocarcinoma, thus representing a possible new target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Simone Polvani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
211
|
The role of mitochondrial electron transport in tumorigenesis and metastasis. Biochim Biophys Acta Gen Subj 2014; 1840:1454-63. [DOI: 10.1016/j.bbagen.2013.10.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 09/20/2013] [Accepted: 10/10/2013] [Indexed: 12/11/2022]
|
212
|
Iqbal MA, Siddiqui FA, Chaman N, Gupta V, Kumar B, Gopinath P, Bamezai RNK. Missense mutations in pyruvate kinase M2 promote cancer metabolism, oxidative endurance, anchorage independence, and tumor growth in a dominant negative manner. J Biol Chem 2014; 289:8098-105. [PMID: 24492614 DOI: 10.1074/jbc.m113.515742] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present study was designed to examine the functional relevance of two heterozygous mutations (H391Y and K422R), observed earlier by us in the Bloom syndrome condition. Cells stably expressing exogenous wild-type or mutant PKM2 (K422R or H391Y) or co-expressing both wild type and mutant (PKM2-K422R or PKM2-H391Y) were assessed for cancer metabolism and tumorigenic potential. Interestingly, cells co-expressing PKM2 and mutant (K422R or H391Y) showed significantly aggressive cancer metabolism as compared with cells expressing either wild-type or mutant PKM2 independently. A similar trend was observed for oxidative endurance, tumorigenic potential, cellular proliferation, and tumor growth. These observations signify the dominant negative nature of mutations. Remarkably, PKM2-H391Y co-expressed cells showed a maximal effect on all the studied parameters. Such a dominant negative impaired function of PKM2 in tumor development is not known; this study demonstrates for the first time the possible predisposition of Bloom syndrome patients with impaired PKM2 activity to cancer and the importance of studying genetic variations in PKM2 in the future to understand their relevance in cancer in general.
Collapse
Affiliation(s)
- Mohd Askandar Iqbal
- From the National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067 India
| | | | | | | | | | | | | |
Collapse
|
213
|
The stem cell transcription factor ZFP57 induces IGF2 expression to promote anchorage-independent growth in cancer cells. Oncogene 2014; 34:752-60. [PMID: 24469060 DOI: 10.1038/onc.2013.599] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 11/25/2013] [Accepted: 12/13/2013] [Indexed: 01/14/2023]
Abstract
Several common biological properties between cancer cells and embryonic stem (ES) cells suggest the possibility that some genes expressed in ES cells might have important roles in cancer cell growth. The transcription factor ZFP57 is expressed in self-renewing ES cells and its expression level decreases during ES cell differentiation. This study showed that ZFP57 is involved in the anchorage-independent growth of human fibrosarcoma HT1080 cells in soft agar. ZFP57 overexpression enhanced, whereas knockdown suppressed, HT1080 tumor formation in nude mice. Furthermore, ZFP57 regulates the expression of insulin-like growth factor 2 (IGF2), which has a critical role in ZFP57-induced anchorage-independent growth. ZFP57 also promotes anchorage-independent growth in ES cells and immortal fibroblasts. Finally, immunohistochemical analysis revealed that ZFP57 is overexpressed in human cancer clinical specimens. Taken together, these results suggest that the ES-specific transcription factor ZFP57 is a novel oncogene.
Collapse
|
214
|
Ahn J, Lee JS, Yang KM. Ultrafine particles of Ulmus davidiana var. japonica induce apoptosis of gastric cancer cells via activation of caspase and endoplasmic reticulum stress. Arch Pharm Res 2014; 37:783-92. [PMID: 24395528 PMCID: PMC4047481 DOI: 10.1007/s12272-013-0312-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 11/24/2013] [Indexed: 01/16/2023]
Abstract
Small-sized particles are more suitable for targeted delivery and are therapeutically more effective than large-sized particles. In this study, we investigated the anticancer effects of ultrafine particles of Ulmus davidiana var. japonica (ufUJ) on human gastric cancer cell lines SNU-1, SNU-216, and SNU-484. ufUJ induced apoptosis by the proteolytic activation of caspase-9, caspase-6, and caspase-3 and cleavage of poly (ADP-ribose) polymerase. The expression levels of the endoplasmic reticulum stress-related protein BiP markedly increased after ufUJ treatment. BiP knockdown decreased ufUJ-induced cell death. ufUJ-induced apoptosis was inhibited by the caspase-3 inhibitor z-DEVD-fmk, caspase-6 inhibitor z-VEID-fmk, and caspase-9 inhibitor z-LEHD-fmk, and by siRNAs against caspases 3, 6, and 9. Gastric cancer cells did not show anchorage-independent growth in the presence of ufUJ. However, cells treated with caspase inhibitors showed an enhanced colony-forming ability. These findings may be helpful in the prevention of gastric cancer and in the development of functional foods.
Collapse
Affiliation(s)
- Joungjwa Ahn
- Department of Food Science and Industry, Jungwon University, Goesan-gun, Chungbuk, Korea
| | | | | |
Collapse
|
215
|
Helfrich I, Ullrich N, Zigrino P, Schadendorf D. Primary tumor versus metastasis: new experimental models for studies on cancer cell homing and metastasis in melanoma. Pigment Cell Melanoma Res 2014; 27:309-16. [PMID: 24314235 DOI: 10.1111/pcmr.12204] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Iris Helfrich
- Skin Caner Unit of the Dermatology Department, University Hospital, University Duisburg-Essen, Duisburg and Essen, Germany
| | | | | | | |
Collapse
|
216
|
Lv Q, Hua F, Hu ZW. Use of the tumor repressor DEDD as a prognostic marker of cancer metastasis. Methods Mol Biol 2014; 1165:197-222. [PMID: 24839027 DOI: 10.1007/978-1-4939-0856-1_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
DEDD, a member of a family of death effector domain-containing proteins, plays crucial roles in mediating apoptosis, regulating cell cycle, and inhibiting cell mitosis. Our recent work demonstrates that DEDD is a novel tumor repressor, which impedes metastasis by reversing the epithelial-mesenchymal transition (EMT) process in breast and colon cancers. DEDD expression therefore may represent a prognostic marker and potential therapeutic target for the prevention and treatment of cancer metastasis. To reveal the anti-metastatic roles of DEDD in these cancer cells, a number of experiments, including immunohistochemistry, the establishment of stably overexpressing or silencing cancer cells, chemoinvasion assay, soft agar assay, protein degradation, and protein-protein interaction were used in our in vitro and in vivo studies. This chapter focuses on the details of these experiments to provide references for the researchers to investigate the function of a gene in the regulation of tumor metastasis.
Collapse
Affiliation(s)
- Qi Lv
- Molecular Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | | | | |
Collapse
|
217
|
Ou C, Zhang J, Shi Y, Wang Z, Wang L, Yang Z, Chen M. d-amino acid doping peptide hydrogel for the production of a cell colony. RSC Adv 2014. [DOI: 10.1039/c3ra46847j] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
218
|
Marampon F, Gravina GL, Popov VM, Scarsella L, Festuccia C, La Verghetta ME, Parente S, Cerasani M, Bruera G, Ficorella C, Ricevuto E, Tombolini V, Di Cesare E, Zani BM. Close correlation between MEK/ERK and Aurora-B signaling pathways in sustaining tumorigenic potential and radioresistance of gynecological cancer cell lines. Int J Oncol 2013; 44:285-94. [PMID: 24189697 DOI: 10.3892/ijo.2013.2167] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/30/2013] [Indexed: 11/06/2022] Open
Abstract
Both Aurora-A and -B kinases have been implicated in tumorigenesis; and as such, they represent an attractive therapeutic target. Recent studies found that Aurora-A is a downstream target of mitogen-activated protein kinase 1/ERK2, while Aurora-B has been found to be a prognostic/predictive therapeutic target for epithelial cancer. In a wide range of human cancers, the Ras/Raf/MEK/ERK/MAP kinase pathway is enhanced and the cellular response to growth signals is known to increase. The purpose of this study was to investigate whether the MEK/ERK cascade regulates tumorigenic signaling and radioresistance via the Aurora-B-mediated pathway in a panel of gynecological cancer cell lines. Exponentially growing human endometrial (Ishikawa), cervical (HeLa), cervical (CASKI) and vulva (SiHa) cancer cells were used in culture treated with either control or MEK/ERK inhibitor or AZD1152 before and after irradiation. Western blotting, ERK1/2 siRNA transfection, growth assay in modified monolayer, Annexin V and migration/invasion assays were performed. The specific MEK/ERK inhibitor U0126 decreased the tumorigenic potential and improved the radiation response in all cellular models. The modulation of radioresponse upon U0126 treatment positively correlated with the inhibition of phospho-ERKs and the reduction of Aurora-B kinase expression. In addition, upon U0126 treatment DNA-PKcs protein expression was found to be downregulated, indicating that the improved radiation response may be caused by decreased DNA double-strand damage repair mechanisms. The knockdown of ERK by siRNA confirmed the MEK/ERK-dependent Aurora-B kinase expression. The use of AZD1152, a selective Aurora-B inhibitor, counteracted tumorigenic potential and radioresistance phenotype by highly increasing apoptotic mechanisms in all gynecological cancer cell lines used. Evidence from our experiments show that tumorigenic potential and radiation response in gynecological cancer cells may ensue from a MEK/ERK or Aurora-B inhibition. Together with the close correlation of MEK/ERK and Aurora-B protein expression, this study underlines the potential role of a MEK/ERK/Aurora-B axis whose interruption recovers the antitumor effects of radiotherapy.
Collapse
Affiliation(s)
- Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, Division of Radiotherapy and Radiobiology Laboratory, San Salvatore Hospital, University of L'Aquila, I-67100 L'Aquila, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Thapa N, Choi S, Hedman A, Tan X, Anderson RA. Phosphatidylinositol phosphate 5-kinase Iγi2 in association with Src controls anchorage-independent growth of tumor cells. J Biol Chem 2013; 288:34707-18. [PMID: 24151076 DOI: 10.1074/jbc.m113.512848] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A fundamental property of tumor cells is to defy anoikis, cell death caused by a lack of cell-matrix interaction, and grow in an anchorage-independent manner. How tumor cells organize signaling molecules at the plasma membrane to sustain oncogenic signals in the absence of cell-matrix interactions remains poorly understood. Here, we describe a role for phosphatidylinositol 4-phosphate 5-kinase (PIPK) Iγi2 in controlling anchorage-independent growth of tumor cells in coordination with the proto-oncogene Src. PIPKIγi2 regulated Src activation downstream of growth factor receptors and integrins. PIPKIγi2 directly interacted with the C-terminal tail of Src and regulated its subcellular localization in concert with talin, a cytoskeletal protein targeted to focal adhesions. Co-expression of PIPKIγi2 and Src synergistically induced the anchorage-independent growth of nonmalignant cells. This study uncovers a novel mechanism where a phosphoinositide-synthesizing enzyme, PIPKIγi2, functions with the proto-oncogene Src, to regulate oncogenic signaling.
Collapse
Affiliation(s)
- Narendra Thapa
- From the School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | | | | | | | | |
Collapse
|
220
|
Sen S, Kawahara B, Fry NL, Farias-Eisner R, Zhang D, Mascharak PK, Chaudhuri G. A light-activated NO donor attenuates anchorage independent growth of cancer cells: Important role of a cross talk between NO and other reactive oxygen species. Arch Biochem Biophys 2013; 540:33-40. [PMID: 24157690 DOI: 10.1016/j.abb.2013.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 10/06/2013] [Accepted: 10/09/2013] [Indexed: 12/12/2022]
Abstract
It is established that high concentrations of nitric oxide(1) (NO), as released from activated macrophages, induce apoptosis in breast cancer cells. In this study, we assessed the potential of a light-activated NO donor [(Me2bpb)Ru(NO)(Resf)], a recently reported apoptototic agent, in suppressing the anchorage independent growth potentials of an aggressive human breast cancer cell line. Our results demonstrated the down regulation of anchorage independent growth by light activated NO treatment in the aggressive human breast cancer cell line MDA-MB-231 and afforded insight into the associated mechanism(s). The investigation revealed an up-regulation of the bioactivity of catalase with an accompanied reduction in the endogenous levels of H2O2, a direct substrate of catalase and a recently identified endogenous growth modulator in breast cancer cells. An earlier publication reported that endogenous superoxide (O2(-)) in human breast cancer cells constitutively inhibits catalase bioactivity (at the level of its protein), resulting in increased H2O2 levels. Interestingly in this study, O2(-) was also found to be down- regulated following NO treatment providing a basis for the observed increase in catalase bioactivity. Cells silenced for the catalase gene exhibited compromised reduction in anchorage independent growth upon light activated NO treatment. Collectively this study detailed a mechanistic cross talk between exogenous NO and endogenous ROS in attenuating anchorage independent growth.
Collapse
Affiliation(s)
- Suvajit Sen
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | |
Collapse
|
221
|
CREB3L1 is a metastasis suppressor that represses expression of genes regulating metastasis, invasion, and angiogenesis. Mol Cell Biol 2013; 33:4985-95. [PMID: 24126059 DOI: 10.1128/mcb.00959-13] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The unfolded protein response (UPR) is activated in response to hypoxia-induced stress such as in the tumor microenvironment. This study examined the role of CREB3L1 (cyclic AMP [cAMP]-responsive element-binding protein 3-like protein 1), a member of the UPR, in breast cancer development and metastasis. Initial experiments identified the loss of CREB3L1 expression in metastatic breast cancer cell lines compared to low-metastasis or nonmetastatic cell lines. When metastatic cells were transfected with CREB3L1, they demonstrated reduced invasion and migration in vitro, as well as a significantly decreased ability to survive under nonadherent or hypoxic conditions. Interestingly, in an in vivo rat mammary tumor model, not only did CREB3L1-expressing cells fail to form metastases compared to CREB3L1 null cells but regression of the primary tumors was seen in 70% of the animals as a result of impaired angiogenesis. Microarray and chromatin immunoprecipitation with microarray technology (ChIP on Chip) analyses identified changes in the expression of many genes involved in cancer development and metastasis, including a decrease in those involved in angiogenesis. These data suggest that CREB3L1 plays an important role in suppressing tumorigenesis and that loss of expression is required for the development of a metastatic phenotype.
Collapse
|
222
|
PAQUIN MARIECHRISTINE, LEBLANC CAROLINE, LEMIEUX ETIENNE, BIAN BENJAMIN, RIVARD NATHALIE. Functional impact of colorectal cancer-associated mutations in the transcription factor E2F4. Int J Oncol 2013; 43:2015-22. [DOI: 10.3892/ijo.2013.2131] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/09/2013] [Indexed: 11/06/2022] Open
|
223
|
Reversal of chemosensitivity and induction of cell malignancy of a non-malignant prostate cancer cell line upon extracellular vesicle exposure. Mol Cancer 2013; 12:118. [PMID: 24103426 PMCID: PMC3851868 DOI: 10.1186/1476-4598-12-118] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/16/2013] [Indexed: 02/03/2023] Open
Abstract
Background Extracellular vesicle (EV) trafficking is a fundamental cellular process that occurs in cells and is required for different aspects of pathophysiology. EV trafficking leads to changes in cellular function including apoptosis, angiogenesis and proliferation required for increased tumor formation. Results We report several phenotypic changes mediated by EVs isolated from non-malignant and malignant prostate cells as well as patient biopsied prostate tumor samples. EVs can reverse the resistance of prostate cancer cells to camptothecin EVs isolated from non-malignant PrECs (Prostate Epithelial Cells) can reverse soft agar colony formation of malignant DU145 cells, with the reciprocal effect observed. Isolation of EVs from 2 Gleason grade 8 prostate cancer patients significantly induced soft agar colony formation of non-malignant PrECs. We have identified proteins via antibody and Mass spectrometry analysis that may be responsible for the phenotypic changes. Mass spectrometry analysis of protein lysates using ProteoIQ revealed protein candidates associated with gene ontology annotations that may be responsible for this phenotypic change. Ingenuity Pathway Analysis was used to identify statistically relevant canonical pathways and functions associated the protein IDs and expression values obtained using ProteoIQ. Western blot analysis confirmed the increase of 14-3-3 zeta, pRKIP and prohibitin protein levels in PrEC cells co-cultured with patient EVs. 14-3-3 proteins were also found as common proteins of 3 other Gleason grade 8 patients. Conclusion Our study provides a rational basis to further investigate putative proteins, such as 14-3-3 and prohibitin and genetic factors that may be responsible for phenotypic changes that are associated with prostate cancer progression.
Collapse
|
224
|
Tan TZ, Miow QH, Huang RYJ, Wong MK, Ye J, Lau JA, Wu MC, Bin Abdul Hadi LH, Soong R, Choolani M, Davidson B, Nesland JM, Wang LZ, Matsumura N, Mandai M, Konishi I, Goh BC, Chang JT, Thiery JP, Mori S. Functional genomics identifies five distinct molecular subtypes with clinical relevance and pathways for growth control in epithelial ovarian cancer. EMBO Mol Med 2013; 5:1051-66. [PMID: 23666744 PMCID: PMC3721473 DOI: 10.1002/emmm.201201823] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 04/03/2013] [Accepted: 04/09/2013] [Indexed: 01/06/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is hallmarked by a high degree of heterogeneity. To address this heterogeneity, a classification scheme was developed based on gene expression patterns of 1538 tumours. Five, biologically distinct subgroups - Epi-A, Epi-B, Mes, Stem-A and Stem-B - exhibited significantly distinct clinicopathological characteristics, deregulated pathways and patient prognoses, and were validated using independent datasets. To identify subtype-specific molecular targets, ovarian cancer cell lines representing these molecular subtypes were screened against a genome-wide shRNA library. Focusing on the poor-prognosis Stem-A subtype, we found that two genes involved in tubulin processing, TUBGCP4 and NAT10, were essential for cell growth, an observation supported by a pathway analysis that also predicted involvement of microtubule-related processes. Furthermore, we observed that Stem-A cell lines were indeed more sensitive to inhibitors of tubulin polymerization, vincristine and vinorelbine, than the other subtypes. This subtyping offers new insights into the development of novel diagnostic and personalized treatment for EOC patients.
Collapse
Affiliation(s)
- Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
| | - Qing Hao Miow
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of SingaporeSingapore
| | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
- Department of Obstetrics and Gynecology, National University Health SystemSingapore
| | - Meng Kang Wong
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
| | - Jieru Ye
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
| | - Jieying Amelia Lau
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
| | - Meng Chu Wu
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
| | | | - Richie Soong
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynecology, National University Health SystemSingapore
| | - Ben Davidson
- Division of Pathology, Norwegian Radium Hospital Oslo University HospitalOslo, Norway
- Faculty of Medicine, University of Oslo, Institute of Clinical MedicineOslo, Norway
| | - Jahn M Nesland
- Division of Pathology, Norwegian Radium Hospital Oslo University HospitalOslo, Norway
- Faculty of Medicine, University of Oslo, Institute of Clinical MedicineOslo, Norway
| | - Ling-Zhi Wang
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
- Department of Pharmacology, National University of SingaporeSingapore
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kyoto UniversityKyoto, Japan
| | - Masaki Mandai
- Department of Obstetrics and Gynecology, Kyoto UniversityKyoto, Japan
| | - Ikuo Konishi
- Department of Obstetrics and Gynecology, Kyoto UniversityKyoto, Japan
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
- Department of Pharmacology, National University of SingaporeSingapore
- Department of Hematology and Oncology, National University Health SystemSingapore
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at HoustonTX, USA
| | - Jean Paul Thiery
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
- Institute of Molecular and Cell Biology, A*STAR (Agency for ScienceTechnology and Research), Singapore
- Department of Biochemistry, National University of SingaporeSingapore
| | - Seiichi Mori
- Cancer Science Institute of Singapore, National University of SingaporeSingapore
- Department of Biochemistry, National University of SingaporeSingapore
- Division of Cancer Genomics, Cancer Institute of Japanese Foundation for Cancer Research3-8-31 Ariake, Koto-ku, Tokyo, Japan
- Present Address: Division of Cancer Genomics, Cancer Institute of Japanese Foundation for Cancer ResearchKoto-ku, Tokyo, Japan
| |
Collapse
|
225
|
Feduska JM, Garcia PL, Brennan SB, Bu S, Council LN, Yoon KJ. N-glycosylation of ICAM-2 is required for ICAM-2-mediated complete suppression of metastatic potential of SK-N-AS neuroblastoma cells. BMC Cancer 2013; 13:261. [PMID: 23714211 PMCID: PMC3700849 DOI: 10.1186/1471-2407-13-261] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 05/22/2013] [Indexed: 11/12/2022] Open
Abstract
Background Cell adhesion molecules (CAMs) are expressed ubiquitously. Each of the four families of CAMs is comprised of glycosylated, membrane-bound proteins that participate in multiple cellular processes including cell-cell communication, cell motility, inside-out and outside-in signaling, tumorigenesis, angiogenesis and metastasis. Intercellular adhesion molecule-2 (ICAM-2), a member of the immunoglobulin superfamily of CAMs, has six N-linked glycosylation sites at amino acids (asparagines) 47, 82, 105, 153, 178 and 187. Recently, we demonstrated a previously unknown function for ICAM-2 in tumor cells. We showed that ICAM-2 suppressed neuroblastoma cell motility and growth in soft agar, and induced a juxtamembrane distribution of F-actin in vitro. We also showed that ICAM-2 completely suppressed development of disseminated tumors in vivo in a murine model of metastatic NB. These effects of ICAM-2 on NB cell phenotype in vitro and in vivo depended on the interaction of ICAM-2 with the cytoskeletal linker protein α-actinin. Interestingly, ICAM-2 did not suppress subcutaneous growth of tumors in mice, suggesting that ICAM-2 affects the metastatic but not the tumorigenic potential of NB cells. The goal of the study presented here was to determine if the glycosylation status of ICAM-2 influenced its function in neuroblastoma cells. Methods Because it is well documented that glycosylation facilitates essential steps in tumor progression and metastasis, we investigated whether the glycosylation status of ICAM-2 affected the phenotype of NB cells. We used site-directed mutagenesis to express hypo- or non-glycosylated variants of ICAM-2, by substituting alanine for asparagine at glycosylation sites, and compared the impact of each variant on NB cell motility, anchorage-independent growth, interaction with intracellular proteins, effect on F-actin distribution and metastatic potential in vivo. Results The in vitro and in vivo phenotypes of cells expressing glycosylation site variants differed from cells expressing fully-glycosylated ICAM-2 or no ICAM-2. Most striking was the finding that mice injected intravenously with NB cells expressing glycosylation site variants survived longer (P ≤ 0.002) than mice receiving SK-N-AS cells with undetectable ICAM-2. However, unlike fully-glycosylated ICAM-2, glycosylation site variants did not completely suppress disseminated tumor development. Conclusions Reduced glycosylation of ICAM-2 significantly attenuated, but did not abolish, its ability to suppress metastatic properties of NB cells.
Collapse
Affiliation(s)
- Joseph M Feduska
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | |
Collapse
|
226
|
Kharma B, Baba T, Mandai M, Matsumura N, Murphy SK, Kang HS, Yamanoi K, Hamanishi J, Yamaguchi K, Yoshioka Y, Konishi I. Utilization of genomic signatures to identify high-efficacy candidate drugs for chemorefractory endometrial cancers. Int J Cancer 2013; 133:2234-44. [DOI: 10.1002/ijc.28220] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/03/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Budiman Kharma
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Susan K. Murphy
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology; Duke University Medical Center; Durham; NC
| | - Hyun Sook Kang
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Yumiko Yoshioka
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics; Kyoto University Graduate School of Medicine; Kyoto; Japan
| |
Collapse
|
227
|
Tan TZ, Miow QH, Huang RYJ, Wong MK, Ye J, Lau JA, Wu MC, Bin Abdul Hadi LH, Soong R, Choolani M, Davidson B, Nesland JM, Wang LZ, Matsumura N, Mandai M, Konishi I, Goh BC, Chang JT, Thiery JP, Mori S. Functional genomics identifies five distinct molecular subtypes with clinical relevance and pathways for growth control in epithelial ovarian cancer. EMBO Mol Med 2013. [PMID: 23666744 DOI: 10.1002/emmm.201201823] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is hallmarked by a high degree of heterogeneity. To address this heterogeneity, a classification scheme was developed based on gene expression patterns of 1538 tumours. Five, biologically distinct subgroups - Epi-A, Epi-B, Mes, Stem-A and Stem-B - exhibited significantly distinct clinicopathological characteristics, deregulated pathways and patient prognoses, and were validated using independent datasets. To identify subtype-specific molecular targets, ovarian cancer cell lines representing these molecular subtypes were screened against a genome-wide shRNA library. Focusing on the poor-prognosis Stem-A subtype, we found that two genes involved in tubulin processing, TUBGCP4 and NAT10, were essential for cell growth, an observation supported by a pathway analysis that also predicted involvement of microtubule-related processes. Furthermore, we observed that Stem-A cell lines were indeed more sensitive to inhibitors of tubulin polymerization, vincristine and vinorelbine, than the other subtypes. This subtyping offers new insights into the development of novel diagnostic and personalized treatment for EOC patients.
Collapse
Affiliation(s)
- Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Relan V, Morrison L, Parsonson K, Clarke BE, Duhig EE, Windsor MN, Matar KS, Naidoo R, Passmore L, McCaul E, Courtney D, Yang IA, Fong KM, Bowman RV. Phenotypes and karyotypes of human malignant mesothelioma cell lines. PLoS One 2013; 8:e58132. [PMID: 23516439 PMCID: PMC3597627 DOI: 10.1371/journal.pone.0058132] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 01/30/2013] [Indexed: 01/19/2023] Open
Abstract
Background Malignant mesothelioma is an aggressive tumour of serosal surfaces most commonly pleura. Characterised cell lines represent a valuable tool to study the biology of mesothelioma. The aim of this study was to develop and biologically characterise six malignant mesothelioma cell lines to evaluate their potential as models of human malignant mesothelioma. Methods Five lines were initiated from pleural biopsies, and one from pleural effusion of patients with histologically proven malignant mesothelioma. Mesothelial origin was assessed by standard morphology, Transmission Electron Microscopy (TEM) and immunocytochemistry. Growth characteristics were assayed using population doubling times. Spectral karyotyping was performed to assess chromosomal abnormalities. Authentication of donor specific derivation was undertaken by DNA fingerprinting using a panel of SNPs. Results Most of cell lines exhibited spindle cell shape, with some retaining stellate shapes. At passage 2 to 6 all lines stained positively for calretinin and cytokeratin 19, and demonstrated capacity for anchorage-independent growth. At passage 4 to 16, doubling times ranged from 30–72 hours, and on spectral karyotyping all lines exhibited numerical chromosomal abnormalities ranging from 41 to 113. Monosomy of chromosomes 8, 14, 22 or 17 was observed in three lines. One line displayed four different karyotypes at passage 8, but only one karyotype at passage 42, and another displayed polyploidy at passage 40 which was not present at early passages. At passages 5–17, TEM showed characteristic features of mesothelioma ultrastructure in all lines including microvilli and tight intercellular junctions. Conclusion These six cell lines exhibit varying cell morphology, a range of doubling times, and show diverse passage-dependent structural chromosomal changes observed in malignant tumours. However they retain characteristic immunocytochemical protein expression profiles of mesothelioma during maintenance in artificial culture systems. These characteristics support their potential as in vitro model systems for studying cellular, molecular and genetic aspects of mesothelioma.
Collapse
Affiliation(s)
- Vandana Relan
- UQ Thoracic Research Centre, School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Ishii G, Hashimoto H, Atsumi N, Hoshino A, Ochiai A. Morphophenotype of floating colonies derived from a single cancer cell has a critical impact on tumor-forming activity. Pathol Int 2013; 63:29-36. [PMID: 23356223 DOI: 10.1111/pin.12018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 11/19/2012] [Indexed: 12/26/2022]
Abstract
The anchorage-independent colony growth of cancer cells is reportedly correlated with the tumor-forming activity; however, the correlation between the morphophenotype of each colony and the tumor-forming activity has not been clarified. To assess this problem, we cultured single A549 cells (human lung adenocarcinoma cell line) in growth medium in individual wells (n = 426) for 14 days under anchorage-independent conditions and analyzed the resulting growth characteristics. The single A549 cells formed various sizes of floating colonies. The proportion of large colonies (>400 μm) was 3.8% and this proportion increased dramatically with the exogenous addition of EGF (21.6%) or HGF (27.6%). Morphologically, the floating colonies could be divided into: (ii) Type A, spheroid colony; and (ii) Type B, dispersed villous colony. The Type B colonies expressed significantly higher levels of epithelial-mesenchymal transition (EMT)-related mRNAs (Snail 1, ZEB 1, and ZEB2) than the Type A colonies. Furthermore, the subcutaneous injection of a single cell-derived colony with a large size and a Type B morphology resulted in more efficient tumor formation. The present results indicated that the morphophenotypes of floating colonies derived from single cancer cells have a critical impact on tumor-forming activity.
Collapse
Affiliation(s)
- Genichiro Ishii
- Division of Pathology, Research Center for Innovative Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.
| | | | | | | | | |
Collapse
|
230
|
Chanvorachote P, Kowitdamrong A, Ruanghirun T, Sritularak B, Mungmee C, Likhitwitayawuid K. Anti-metastatic Activities of Bibenzyls from Dendrobium pulchellum. Nat Prod Commun 2013. [DOI: 10.1177/1934578x1300800127] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Our investigation of the stem of Dendrobium pulchellum resulted in the isolation of four known bibenzyls, chrysotobibenzyl (1), chrysotoxine (2), crepidatin (3) and moscatilin (4). The present study reveals for the first time the ability of these four compounds to facilitate anoikis and inhibit the growth of lung cancer cells in anchorage-independent condition. The preliminary data obtained disclose the inhibitory effect on cancer cell metastasis of the isolated compounds, and provide an important new approach for cancer drug development.
Collapse
Affiliation(s)
- Pithi Chanvorachote
- Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Thidarat Ruanghirun
- Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Boonchoo Sritularak
- Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chutichot Mungmee
- Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | |
Collapse
|
231
|
Castro IC, Breiling A, Luetkenhaus K, Ceteci F, Hausmann S, Kress S, Lyko F, Rudel T, Rapp UR. MYC-induced epigenetic activation of GATA4 in lung adenocarcinoma. Mol Cancer Res 2012; 11:161-72. [PMID: 23239811 DOI: 10.1158/1541-7786.mcr-12-0414-t] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human lung cancer is a disease with high incidence and accounts for most cancer-related deaths in both men and women. Metastasis is a common event in non-small cell lung carcinoma (NSCLC), diminishing the survival chance of the patients with this type of tumor. It has been shown that MYC is involved in the development of metastasis from NSCLC, but the mechanism underlying this switch remained to be identified. Here, we focus on GATA4 as a MYC target in the development of metastasis with origin in lung adenocarcinoma, the most common type of NSCLC. Epigenetic alterations at the GATA4 promoter level were observed after MYC expression in lung adenocarcinoma in vivo and in vitro. Such alterations include site-specific demethylation that accompanies the displacement of the MYC-associated zinc finger protein (MAZ) from the GATA4 promoter, which leads to GATA4 expression. Histone modification analysis of the GATA4 promoter revealed a switch from repressive histone marks to active histone marks after MYC binding, which corresponds to active GATA4 expression. Our results thus identify a novel epigenetic mechanism by which MYC activates GATA4 leading to metastasis in lung adenocarcinoma, suggesting novel potential targets for the development of antimetastatic therapy.
Collapse
Affiliation(s)
- Inês C Castro
- Biocenter, Department of Microbiology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Deakin NO, Pignatelli J, Turner CE. Diverse roles for the paxillin family of proteins in cancer. Genes Cancer 2012; 3:362-70. [PMID: 23226574 DOI: 10.1177/1947601912458582] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The paxillin family of intracellular scaffold proteins includes paxillin, Hic-5, and leupaxin, and all have been identified as key regulators of the cellular migration machinery in both 2- and 3-dimensional microenvironments. Herein, we provide insight into the roles of these proteins during tumorigenesis and metastasis, highlighting their functions in cancer initiation as well as tumor cell dissemination and survival. Furthermore, we speculate on the potential of paxillin family proteins as both future prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Nicholas O Deakin
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | | | | |
Collapse
|
233
|
Clocchiatti A, Di Giorgio E, Ingrao S, Meyer-Almes FJ, Tripodo C, Brancolini C. Class IIa HDACs repressive activities on MEF2-depedent transcription are associated with poor prognosis of ER⁺ breast tumors. FASEB J 2012; 27:942-54. [PMID: 23159930 DOI: 10.1096/fj.12-209346] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
MEF2s transcription factors and class IIa HDACs compose a fundamental axis for several differentiation pathways. Functional relationships between this axis and cancer are largely unexplored. We have found that class IIa HDACs are heterogeneously expressed and display redundant activities in breast cancer cells. Applying gene set enrichment analysis to compare the expression profile of a list of putative MEF2 target genes, we have discovered a correlation between the down-regulation of the MEF2 signature and the aggressiveness of ER(+) breast tumors. Kaplan-Meier analysis in ER(+) breast tumors evidenced an association between increased class IIa HDACs expression and reduced survival. The important role of the MEF2-HDAC axis in ER(+) breast cancer was confirmed in cultured cells. MCF7 ER(+) cells were susceptible to silencing of class IIa HDACs in terms of both MEF2-dependent transcription and apoptosis. Conversely, in ER(-) MDA-MB-231 cells, the repressive influence of class IIa HDACs was dispensable. Similarly, a class IIa HDAC-specific inhibitor preferentially promoted the up-regulation of several MEF2 target genes and apoptosis in ER(+) cell lines. The prosurvival function of class IIa HDACs could be explained by the repression of NR4A1/Nur77, a proapoptotic MEF2 target. In summary, our studies underscore a contribution of class IIa HDACs to aggressiveness of ER(+) tumors.-Clocchiatti, A., Di Giorgio, E., Ingrao, S., Meyer-Almes, F.-J., Tripodo, C., Brancolini, C. Class IIa HDACs repressive activities on MEF2-depedent transcription are associated with poor prognosis of ER(+) breast tumors.
Collapse
Affiliation(s)
- Andrea Clocchiatti
- Dipartimento di Scienze Mediche e Biologiche Universita' di Udine. P.le Kolbe 4-33100, Udine, Italy
| | | | | | | | | | | |
Collapse
|
234
|
Long-term hydrogen peroxide exposure potentiates anoikis resistance and anchorage-independent growth in lung carcinoma cells. Cell Biol Int 2012; 36:1055-66. [DOI: 10.1042/cbi20120111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
235
|
Sampson ER, McMurray HR, Hassane DC, Newman L, Salzman P, Jordan CT, Land H. Gene signature critical to cancer phenotype as a paradigm for anticancer drug discovery. Oncogene 2012; 32:3809-18. [PMID: 22964631 DOI: 10.1038/onc.2012.389] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 04/25/2012] [Accepted: 07/20/2012] [Indexed: 02/06/2023]
Abstract
Malignant cell transformation commonly results in the deregulation of thousands of cellular genes, an observation that suggests a complex biological process and an inherently challenging scenario for the development of effective cancer interventions. To better define the genes/pathways essential to regulating the malignant phenotype, we recently described a novel strategy based on the cooperative nature of carcinogenesis that focuses on genes synergistically deregulated in response to cooperating oncogenic mutations. These so-called 'cooperation response genes' (CRGs) are highly enriched for genes critical for the cancer phenotype, thereby suggesting their causal role in the malignant state. Here, we show that CRGs have an essential role in drug-mediated anticancer activity and that anticancer agents can be identified through their ability to antagonize the CRG expression profile. These findings provide proof-of-concept for the use of the CRG signature as a novel means of drug discovery with relevance to underlying anticancer drug mechanisms.
Collapse
Affiliation(s)
- E R Sampson
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
236
|
Zhang L, Komurov K, Wright WE, Shay JW. Identification of novel driver tumor suppressors through functional interrogation of putative passenger mutations in colorectal cancer. Int J Cancer 2012; 132:732-7. [PMID: 22753261 DOI: 10.1002/ijc.27705] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 06/11/2012] [Indexed: 01/23/2023]
Abstract
Cancer genome sequencing efforts are leading to the identification of genetic mutations in many types of malignancy. However, the majority of these genetic alterations have been considered random passengers that do not directly contribute to tumorigenesis. We have previously conducted a soft agar-based short hairpin RNA (shRNA) screen within colorectal cancer (CRC) candidate driver genes (CAN-genes) using a karyotypically diploid hTERT- and CDK4-immortalized human colonic epithelial cell (HCEC) model and discovered that depletion of 65 of the 151 CAN-genes enhanced anchorage-independent growth in HCECs with ectopic expression of K-Ras(V12) and/or TP53 knockdown. We now constructed an interaction map of the confirmed CAN-genes with CRC non-CAN-genes and screened for functional tumor suppressors. Remarkably, depletion of 15 out of 25 presumed passenger genes that interact with confirmed CAN-genes (60%) promoted soft agar growth in HCECs with TP53 knockdown compared to only 7 out of 55 (12.5%) of presumed passenger genes that do not interact. We have thus demonstrated a pool of driver mutations among the putative CRC passenger/incidental mutations, establishing the importance of employing biological filters, in addition to bioinformatics, to identify driver mutations.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9039, USA
| | | | | | | |
Collapse
|
237
|
The Interactions between Insulin and Androgens in Progression to Castrate-Resistant Prostate Cancer. Adv Urol 2012; 2012:248607. [PMID: 22548055 PMCID: PMC3324133 DOI: 10.1155/2012/248607] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 01/06/2012] [Indexed: 12/31/2022] Open
Abstract
An association between the metabolic syndrome and reduced testosterone levels has been identified, and a specific inverse relationship between insulin and testosterone levels suggests that an important metabolic crosstalk exists between these two hormonal axes; however, the mechanisms by which insulin and androgens may be reciprocally regulated are not well described. Androgen-dependant gene pathways regulate the growth and maintenance of both normal and malignant prostate tissue, and androgen-deprivation therapy (ADT) in patients exploits this dependence when used to treat recurrent and metastatic prostate cancer resulting in tumour regression. A major systemic side effect of ADT includes induction of key features of the metabolic syndrome and the consistent feature of hyperinsulinaemia. Recent studies have specifically identified a correlation between elevated insulin and high-grade PCa and more rapid progression to castrate resistant disease. This paper examines the relationship between insulin and androgens in the context of prostate cancer progression. Prostate cancer patients present a promising cohort for the exploration of insulin stabilising agents as adjunct treatments for hormone deprivation or enhancers of chemosensitivity for treatment of advanced prostate cancer.
Collapse
|
238
|
Donigan KA, Hile SE, Eckert KA, Sweasy JB. The human gastric cancer-associated DNA polymerase β variant D160N is a mutator that induces cellular transformation. DNA Repair (Amst) 2012; 11:381-90. [PMID: 22341651 DOI: 10.1016/j.dnarep.2012.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/04/2012] [Accepted: 01/05/2012] [Indexed: 10/28/2022]
Abstract
Approximately 30% of human tumors sequenced to date harbor mutations in the POLB gene that are not present in matched normal tissue. Many mutations give rise to enzymes that contain non-synonymous single amino acid substitutions, several of which have been found to have aberrant activity or fidelity and transform cells when expressed. The DNA Polymerase β (Pol β) variant Asp160Asn (D160N) was first identified in a gastric tumor. Expression of D160N in cells induces cellular transformation as measured by hyperproliferation, focus formation, anchorage-independent growth and invasion. Here, we show that D160N is an active mutator polymerase that induces complex mutations. Our data support the interpretation that complex mutagenesis is the underlying mechanism of the observed cellular phenotypes, all of which are linked to tumorigenesis or tumor progression.
Collapse
Affiliation(s)
- Katherine A Donigan
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520, United States
| | | | | | | |
Collapse
|
239
|
Venet D, Dumont JE, Detours V. Most random gene expression signatures are significantly associated with breast cancer outcome. PLoS Comput Biol 2011; 7:e1002240. [PMID: 22028643 PMCID: PMC3197658 DOI: 10.1371/journal.pcbi.1002240] [Citation(s) in RCA: 438] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 09/07/2011] [Indexed: 12/19/2022] Open
Abstract
Bridging the gap between animal or in vitro models and human disease is essential in medical research. Researchers often suggest that a biological mechanism is relevant to human cancer from the statistical association of a gene expression marker (a signature) of this mechanism, that was discovered in an experimental system, with disease outcome in humans. We examined this argument for breast cancer. Surprisingly, we found that gene expression signatures—unrelated to cancer—of the effect of postprandial laughter, of mice social defeat and of skin fibroblast localization were all significantly associated with breast cancer outcome. We next compared 47 published breast cancer outcome signatures to signatures made of random genes. Twenty-eight of them (60%) were not significantly better outcome predictors than random signatures of identical size and 11 (23%) were worst predictors than the median random signature. More than 90% of random signatures >100 genes were significant outcome predictors. We next derived a metagene, called meta-PCNA, by selecting the 1% genes most positively correlated with proliferation marker PCNA in a compendium of normal tissues expression. Adjusting breast cancer expression data for meta-PCNA abrogated almost entirely the outcome association of published and random signatures. We also found that, in the absence of adjustment, the hazard ratio of outcome association of a signature strongly correlated with meta-PCNA (R2 = 0.9). This relation also applied to single-gene expression markers. Moreover, >50% of the breast cancer transcriptome was correlated with meta-PCNA. A corollary was that purging cell cycle genes out of a signature failed to rule out the confounding effect of proliferation. Hence, it is questionable to suggest that a mechanism is relevant to human breast cancer from the finding that a gene expression marker for this mechanism predicts human breast cancer outcome, because most markers do. The methods we present help to overcome this problem. Proving that research findings from in vitro or animal models are relevant to human diseases is a major bottleneck in medical science. Hundreds of researchers have suggested the human relevance of oncogenic mechanisms from the statistical association between gene expression markers of these mechanisms and disease outcome. Such evidence has become easier to obtain recently with the advent of microarray screens and of large public-domain genome-wide expression datasets with patient follow-up. We demonstrated that in breast cancer any set of 100 genes or more selected at random has a 90% chance to be significantly associated with outcome. Thus, investigators are bound to find an association however whimsical their marker is. For example, we could establish outcome associations for a signature of postprandial laughter and a signature of social defeat in mice. Association was not stronger than expected at random for 28 (60%) of 47 published breast cancer signatures. The odds of association are 5–17% with random single gene markers—a finding relevant to older breast cancer studies. We explained these results by showing that much of the breast cancer transcriptome is correlated with proliferation, which integrates most prognostic information in this disease.
Collapse
Affiliation(s)
- David Venet
- IRIDIA-CoDE, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Jacques E. Dumont
- IRIBHM, Université Libre de Bruxelles (U.L.B.), Campus Erasme, Brussels, Belgium
| | - Vincent Detours
- IRIBHM, Université Libre de Bruxelles (U.L.B.), Campus Erasme, Brussels, Belgium
- WELBIO, Université Libre de Bruxelles (U.L.B.), Campus Erasme, Brussels, Belgium
- * E-mail:
| |
Collapse
|
240
|
Tzatsos A, Paskaleva P, Lymperi S, Contino G, Stoykova S, Chen Z, Wong KK, Bardeesy N. Lysine-specific demethylase 2B (KDM2B)-let-7-enhancer of zester homolog 2 (EZH2) pathway regulates cell cycle progression and senescence in primary cells. J Biol Chem 2011; 286:33061-9. [PMID: 21757686 PMCID: PMC3190920 DOI: 10.1074/jbc.m111.257667] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 07/07/2011] [Indexed: 12/13/2022] Open
Abstract
Sustained expression of the histone demethylase, KDM2B (Ndy1/FBXL10/JHDM1B), bypasses cellular senescence in primary mouse embryonic fibroblasts (MEFs). Here, we show that KDM2B is a conserved regulator of lifespan in multiple primary cell types and defines a program in which this chromatin-modifying enzyme counteracts the senescence-associated down-regulation of the EZH2 histone methyltransferase. Senescence in MEFs epigenetically silences KDM2B and induces the tumor suppressor miRNAs let-7b and miR-101, which target EZH2. Forced expression of KDM2B promotes immortalization by silencing these miRNAs through locus-specific histone H3 K36me2 demethylation, leading to EZH2 up-regulation. Overexpression of let-7b down-regulates EZH2, induces premature senescence, and counteracts immortalization of MEFs driven by KDM2B. The KDM2B-let-7-EZH2 pathway also contributes to the proliferation of immortal Ink4a/Arf null fibroblasts suggesting that, beyond its anti-senescence role in primary cells, this histone-modifying enzyme functions more broadly in the regulation of cellular proliferation.
Collapse
Affiliation(s)
| | | | - Stephania Lymperi
- Center for Regenerative Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114 and
| | | | | | - Zhao Chen
- the Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115
| | - Kwok-Kin Wong
- the Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts 02115
| | | |
Collapse
|
241
|
Serganova I, Rizwan A, Ni X, Thakur SB, Vider J, Russell J, Blasberg R, Koutcher JA. Metabolic imaging: a link between lactate dehydrogenase A, lactate, and tumor phenotype. Clin Cancer Res 2011; 17:6250-6261. [PMID: 21844011 DOI: 10.1158/1078-0432.ccr-11-0397] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE We compared the metabolic profiles and the association between LDH-A expression and lactate production in two isogenic murine breast cancer cell lines and tumors (67NR and 4T1). These cell lines were derived from a single mammary tumor and have different growth and metabolic phenotypes. EXPERIMENTAL DESIGN LDH-A expression, lactate concentration, glucose utilization, and oxygen consumption were measured in cells, and the potential relationship between tumor lactate levels [measured by magnetic resonance spectroscopic imaging (MRSI)] and tumor glucose utilization [measured by [(18)F]2-deoxy-2-fluoro-D-glucose positron emission tomography ([(18)F]FDG-PET)] was assessed in orthotopic breast tumors derived from these cell lines. RESULTS We show a substantial difference in LDH-A expression between 67NR and 4T1 cells under normoxia and hypoxia. We also show that small orthotopic 4T1 tumors generate 10-fold more lactate than corresponding 67NR tumors. The high lactate levels in small primary 4T1 tumors are associated with intense pimonidazole staining (a hypoxia indicator). Less-intense hypoxia staining was observed in the larger 67NR tumors and is consistent with the gradual increase and plateau of lactate concentration in enlarging 67NR tumors. CONCLUSIONS Lactate-MRSI has a greater dynamic range than [(18)F]FDG-PET and may be a more sensitive measure with which to evaluate the aggressive and metastatic potential of primary breast tumors.
Collapse
Affiliation(s)
- Inna Serganova
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| | - Asif Rizwan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| | - Xiaohui Ni
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| | - Sunitha B Thakur
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| | - Jelena Vider
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| | - James Russell
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| | - Ronald Blasberg
- Department of Neurology, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA.,Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| | - Jason A Koutcher
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA.,Department of Radiology, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA.,Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 415 E68 Street, New York, NY 10065, USA
| |
Collapse
|
242
|
Bee A, Brewer D, Beesley C, Dodson A, Forootan S, Dickinson T, Gerard P, Lane B, Yao S, Cooper CS, Djamgoz MBA, Gosden CM, Ke Y, Foster CS. siRNA knockdown of ribosomal protein gene RPL19 abrogates the aggressive phenotype of human prostate cancer. PLoS One 2011; 6:e22672. [PMID: 21799931 PMCID: PMC3142177 DOI: 10.1371/journal.pone.0022672] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 07/04/2011] [Indexed: 12/15/2022] Open
Abstract
We provide novel functional data that posttranscriptional silencing of gene RPL19 using RNAi not only abrogates the malignant phenotype of PC-3M prostate cancer cells but is selective with respect to transcription and translation of other genes. Reducing RPL19 transcription modulates a subset of genes, evidenced by gene expression array analysis and Western blotting, but does not compromise cell proliferation or apoptosis in-vitro. However, growth of xenografted tumors containing the knocked-down RPL19 in-vivo is significantly reduced. Analysis of the modulated genes reveals induction of the non-malignant phenotype principally to involve perturbation of networks of transcription factors and cellular adhesion genes. The data provide evidence that extra-ribosomal regulatory functions of RPL19, beyond protein synthesis, are critical regulators of cellular phenotype. Targeting key members of affected networks identified by gene expression analysis raises the possibility of therapeutically stabilizing a benign phenotype generated by modulating the expression of an individual gene and thereafter constraining a malignant phenotype while leaving non-malignant tissues unaffected.
Collapse
Affiliation(s)
- Alix Bee
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Daniel Brewer
- Molecular Carcinogenesis Group, Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Carol Beesley
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Andrew Dodson
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Shiva Forootan
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Timothy Dickinson
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Patricia Gerard
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Brian Lane
- Liverpool Microarray Facility, Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Sheng Yao
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Colin S. Cooper
- Molecular Carcinogenesis Group, Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Mustafa B. A. Djamgoz
- Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom
| | - Christine M. Gosden
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Youqiang Ke
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Christopher S. Foster
- Section of Cellular Pathology and Molecular Genetics, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
243
|
p140Cap suppresses the invasive properties of highly metastatic MTLn3-EGFR cells via impaired cortactin phosphorylation. Oncogene 2011; 31:624-33. [DOI: 10.1038/onc.2011.257] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
244
|
Xu Y, Duanmu H, Chang Z, Zhang S, Li Z, Li Z, Liu Y, Li K, Qiu F, Li X. The application of gene co-expression network reconstruction based on CNVs and gene expression microarray data in breast cancer. Mol Biol Rep 2011; 39:1627-37. [PMID: 21611746 DOI: 10.1007/s11033-011-0902-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 05/17/2011] [Indexed: 12/20/2022]
Abstract
Copy number variations (CNVs) are one type of the human genetic variations and are pervasive in the human genome. It has been confirmed that they can play a causal role in complex diseases. Previous studies of CNVs focused more on identifying the disease-specific CNV regions or candidate genes on these CNV regions, but less on the synergistic actions between genes on CNV regions and other genes. Our research combined the CNVs with related gene co-expression to reconstruct gene co-expression network by using single nucleotide polymorphism microarray datasets and gene microarray datasets of breast cancer, and then extracted the modules which connected densely inside and analyzed the functions of modules. Interestingly, all of these modules' functions were related to breast cancer according to our enrichment analysis, and most of the genes in these modules have been reported to be involved in breast cancer. Our findings suggested that integrating CNVs and gene co-expressed relations was an available way to analyze the roles of CNV genes and their synergistic genes in breast cancer, and provided a novel insight into the pathological mechanism of breast cancer.
Collapse
Affiliation(s)
- Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Yamamura S, Matsumura N, Mandai M, Huang Z, Oura T, Baba T, Hamanishi J, Yamaguchi K, Kang HS, Okamoto T, Abiko K, Mori S, Murphy SK, Konishi I. The activated transforming growth factor-beta signaling pathway in peritoneal metastases is a potential therapeutic target in ovarian cancer. Int J Cancer 2011; 130:20-8. [PMID: 21503873 DOI: 10.1002/ijc.25961] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 12/20/2010] [Indexed: 11/07/2022]
Abstract
Peritoneal dissemination including omental metastasis is the most frequent route of metastasis and an important prognostic factor in advanced ovarian cancer. We analyzed the publicly available microarray dataset (GSE2109) using binary regression and found that the transforming growth factor (TGF)-beta signaling pathway was activated in omental metastases as compared to primary sites of disease. Immunohistochemical analysis of TGF-beta receptor type 2 and phosphorylated SMAD2 indicated that both were upregulated in omental metastases as compared to primary disease sites. Treatment of the mouse ovarian cancer cell line HM-1 with recombinant TGF-β1 promoted invasiveness, cell motility and cell attachment while these were suppressed by treatment with A-83-01, an inhibitor of the TGF-β signaling pathway. Microarray analysis of HM-1 cells treated with TGF-β1 and/or A-83-01 revealed that A-83-01 efficiently inhibited transcriptional changes that are induced by TGF-β1. Using gene set enrichment analysis, we found that genes upregulated by TGF-β1 in HM-1 cells were also significantly upregulated in omental metastases compared to primary sites in the human ovarian cancer dataset, GSE2109 (false discovery rate (FDR) q = 0.086). Therapeutic effects of A-83-01 in a mouse model of peritoneal dissemination were examined. Intraperitoneal injection of A-83-01 (150 μg given three times weekly) significantly improved survival (p = 0.015). In summary, these results show that the activated TGF-β signaling pathway in peritoneal metastases is a potential therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Shogo Yamamura
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Ooki A, Yamashita K, Kikuchi S, Sakuramoto S, Katada N, Waraya M, Kawamata H, Nishimiya H, Nakamura K, Watanabe M. Therapeutic potential of PRL-3 targeting and clinical significance of PRL-3 genomic amplification in gastric cancer. BMC Cancer 2011; 11:122. [PMID: 21466710 PMCID: PMC3080833 DOI: 10.1186/1471-2407-11-122] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Accepted: 04/06/2011] [Indexed: 12/29/2022] Open
Abstract
Background Phosphatase of regenerating liver-3 (PRL-3) has deserved attention as a crucial molecule in the multiple steps of metastasis. In the present study, we examined the mechanisms regulating PRL-3 expression, and assessed the clinical potential of PRL-3-targeted therapy in gastric cancer. Methods PRL-3 genomic amplification was analyzed using quantitative-polymerase chain reaction and/or fluorescence in situ hybridization in 77 primary gastric tumors. The anticancer activity of PRL-3 inhibitor (1-4-bromo-2-benzylidene rhodanine) treatment was evaluated against cancer cells with different genetic and expression status. Results PRL-3 genomic amplification was closely concordant with high level of its protein expression in cell lines, and was found in 20% (8/40) among human primary tumors with its expression, which were all stage III/IV disease (40%, 8/20), but in none (0/37) among those without expression. Additionally, PRL-3 genomic amplification was associated with metastatic lymph node status, leading to advanced stage and thereby poor outcomes in patients with lymph node metastasis (P = 0.021). PRL-3 small interfering RNA robustly repressed metastatic properties, including cell proliferation, invasion, and anchorage-independent colony formation. Although neither PRL-3 genomic amplification nor expression level was responsible for the sensitivity to PRL-3 inhibitor treatment, the inhibitor showed dose-dependent anticancer efficacy, and remarkably induced apoptosis on all the tested cell lines with PRL-3 expression. Conclusions We have for the first time, demonstrated that PRL-3 genomic amplification is one of the predominant mechanisms inducing its expression, especially in more advanced stage, and that PRL-3-targeted therapy may have a great potential against gastric cancer with its expression.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Surgery, Kitasato University Hospital, Kitasato 1-15-1, Sagamihara 228-8555, Kanagawa, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Symonds JM, Ohm AM, Carter CJ, Heasley LE, Aly TA, Franklin WA, Reyland ME. Protein kinase C δ is a downstream effector of oncogenic K-ras in lung tumors. Cancer Res 2011; 71:2087-97. [PMID: 21335545 PMCID: PMC3271733 DOI: 10.1158/0008-5472.can-10-1511] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oncogenic activation of K-ras occurs commonly in non-small cell lung cancer (NSCLC), but strategies to therapeutically target this pathway have been challenging to develop. Information about downstream effectors of K-ras remains incomplete, and tractable targets are yet to be defined. In this study, we investigated the role of protein kinase C δ (PKCδ) in K-ras-dependent lung tumorigenesis by using a mouse carcinogen model and human NSCLC cells. The incidence of urethane-induced lung tumors was decreased by 69% in PKCδ-deficient knockout (δKO) mice compared with wild-type (δWT) mice. δKO tumors are smaller and showed reduced proliferation. DNA sequencing indicated that all δWT tumors had activating mutations in KRAS, whereas only 69% of δKO tumors did, suggesting that PKCδ acts as a tumor promoter downstream of oncogenic K-ras while acting as a tumor suppressor in other oncogenic contexts. Similar results were obtained in a panel of NSCLC cell lines with oncogenic K-ras but which differ in their dependence on K-ras for survival. RNA interference-mediated attenuation of PKCδ inhibited anchorage-independent growth, invasion, migration, and tumorigenesis in K-ras-dependent cells. These effects were associated with suppression of mitogen-activated protein kinase pathway activation. In contrast, PKCδ attenuation enhanced anchorage-independent growth, invasion, and migration in NSCLC cells that were either K-ras-independent or that had WT KRAS. Unexpectedly, our studies indicate that the function of PKCδ in tumor cells depends on a specific oncogenic context, as loss of PKCδ in NSCLC cells suppressed transformed growth only in cells dependent on oncogenic K-ras for proliferation and survival.
Collapse
Affiliation(s)
- Jennifer M. Symonds
- Program in Cancer Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
| | - Angela M. Ohm
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
| | - Cristan J. Carter
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
| | - Lynn E. Heasley
- Program in Cancer Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
| | - Theresa A. Aly
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
| | - Wilbur A. Franklin
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
| | - Mary E. Reyland
- Program in Cancer Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045
| |
Collapse
|
248
|
Shlomi T, Benyamini T, Gottlieb E, Sharan R, Ruppin E. Genome-scale metabolic modeling elucidates the role of proliferative adaptation in causing the Warburg effect. PLoS Comput Biol 2011; 7:e1002018. [PMID: 21423717 PMCID: PMC3053319 DOI: 10.1371/journal.pcbi.1002018] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 01/28/2011] [Indexed: 12/29/2022] Open
Abstract
The Warburg effect--a classical hallmark of cancer metabolism--is a counter-intuitive phenomenon in which rapidly proliferating cancer cells resort to inefficient ATP production via glycolysis leading to lactate secretion, instead of relying primarily on more efficient energy production through mitochondrial oxidative phosphorylation, as most normal cells do. The causes for the Warburg effect have remained a subject of considerable controversy since its discovery over 80 years ago, with several competing hypotheses. Here, utilizing a genome-scale human metabolic network model accounting for stoichiometric and enzyme solvent capacity considerations, we show that the Warburg effect is a direct consequence of the metabolic adaptation of cancer cells to increase biomass production rate. The analysis is shown to accurately capture a three phase metabolic behavior that is observed experimentally during oncogenic progression, as well as a prominent characteristic of cancer cells involving their preference for glutamine uptake over other amino acids.
Collapse
Affiliation(s)
- Tomer Shlomi
- Computer Science Department, Technion - Israel Institute of Technology, Haifa, Israel
- * E-mail: (TS); (ER)
| | - Tomer Benyamini
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Eyal Gottlieb
- The Beatson Institute for Cancer Research, Glasgow, United Kingdom
| | - Roded Sharan
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Ruppin
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- * E-mail: (TS); (ER)
| |
Collapse
|
249
|
Kang HS, Baba T, Mandai M, Matsumura N, Hamanishi J, Kharma B, Kondoh E, Yoshioka Y, Oishi S, Fujii N, Murphy SK, Konishi I. GPR54 Is a Target for Suppression of Metastasis in Endometrial Cancer. Mol Cancer Ther 2011; 10:580-90. [DOI: 10.1158/1535-7163.mct-10-0763] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
250
|
Ke XS, Li WC, Hovland R, Qu Y, Liu RH, McCormack E, Thorsen F, Olsen JR, Molven A, Kogan-Sakin I, Rotter V, Akslen LA, Oyan AM, Kalland KH. Reprogramming of cell junction modules during stepwise epithelial to mesenchymal transition and accumulation of malignant features in vitro in a prostate cell model. Exp Cell Res 2010; 317:234-47. [PMID: 20969863 DOI: 10.1016/j.yexcr.2010.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 09/28/2010] [Accepted: 10/09/2010] [Indexed: 12/23/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is pivotal in tumor metastasis. Our previous work reported an EMT model based on primary prostate epithelial cells (EP156T) which gave rise to cells with mesenchymal phenotype (EPT1) without malignant transformation. To promote prostate cell transformation, cells were maintained in saturation density cultures to select for cells overriding quiescence. Foci formed repeatedly following around 8 weeks in confluent EPT1 monolayers. Only later passage EPT1, but not EP156T cells of any passage, could form foci. Cells isolated from the foci were named EPT2 and formed robust colonies in soft agar, a malignant feature present neither in EP156T nor in EPT1 cells. EPT2 cells showed additional malignant traits in vitro, including higher ability to proliferate following confluence, higher resistance to apoptosis and lower dependence on exogenous growth factors than EP156T and EPT1 cells. Microarray profiling identified gene sets, many of which belong to cell junction modules, that changed expression from EP156T to EPT1 cells and continued to change from EPT1 to EPT2 cells. Our findings provide a novel stepwise cell culture model in which EMT emerges independently of transformation and is associated with subsequent accumulation of malignant features in prostate cells. Reprogramming of cell junction modules is involved in both steps.
Collapse
Affiliation(s)
- Xi-song Ke
- The Gade Institute, University of Bergen, Bergen, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|