151
|
Yacoubian TA, Slone SR, Harrington AJ, Hamamichi S, Schieltz JM, Caldwell KA, Caldwell GA, Standaert DG. Differential neuroprotective effects of 14-3-3 proteins in models of Parkinson's disease. Cell Death Dis 2010; 1:e2. [PMID: 21152247 PMCID: PMC2998343 DOI: 10.1038/cddis.2009.4] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 10/02/2009] [Indexed: 12/21/2022]
Abstract
14-3-3 proteins are important negative regulators of cell death pathways. Recent studies have revealed alterations in 14-3-3s in Parkinson's disease (PD) and the ability of 14-3-3s to interact with alpha-synuclein (α-syn), a protein central to PD pathophysiology. In a transgenic α-syn mouse model, we found reduced expression of 14-3-3θ, ε, and γ. These same isoforms prevent α-syn inclusion formation in an H4 neuroglioma cell model. Using dopaminergic cell lines stably overexpressing each 14-3-3 isoform, we found that overexpression of 14-3-3θ, ε, or γ led to resistance to both rotenone and 1-methyl-4-phenylpyridinium (MPP(+)), while other isoforms were not protective against both toxins. Inhibition of a single protective isoform, 14-3-3θ, by shRNA did not increase vulnerability to neurotoxic injury, but toxicity was enhanced by broad-based inhibition of 14-3-3 action with the peptide inhibitor difopein. Using a transgenic C. elegans model of PD, we confirmed the ability of both human 14-3-3θ and a C. elegans 14-3-3 homolog (ftt-2) to protect dopaminergic neurons from α-syn toxicity. Collectively, these data show a strong neuroprotective effect of enhanced 14-3-3 expression - particularly of the 14-3-3θ, ε, and γ isoforms - in multiple cellular and animal models of PD, and point to the potential value of these proteins in the development of neuroprotective therapies for human PD.
Collapse
Affiliation(s)
- T A Yacoubian
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | | | | | | | | | | | | |
Collapse
|
152
|
Kioulafa M, Balkouranidou I, Sotiropoulou G, Kaklamanis L, Mavroudis D, Georgoulias V, Lianidou ES. Methylation of cystatin M promoter is associated with unfavorable prognosis in operable breast cancer. Int J Cancer 2009; 125:2887-92. [PMID: 19551853 DOI: 10.1002/ijc.24686] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The methylation status of cystatin M (CST6) gene in breast tumors was investigated and its prognostic significance as a novel breast cancer biomarker was evaluated. Using methylation-specific PCR (MSP), CST6 promoter methylation was examined in 134 formalin fixed paraffin-embedded tissues (FFPEs): 10 pairs of breast tumors and their surrounding normal tissues, 10 breast fibroadenomas, 11 normal breast tissues and 93 breast tumors. Methylation of CST6 promoter was observed in 2/21 (9.5%) noncancerous breast tissues, 1/10 (10%) benign breast tumors (fibroadenomas) and 52 (55.9%) operable breast cancer tumor samples. CST6 was rarely methylated in the normal tissue surrounding the tumor (10%). During the follow-up period, 24 (25.8%) patients relapsed and 19 (20.4%) died. CST6 methylation was detected in 19 (79.2%) of patients who relapsed and in 15 (78.9%) of patients who died. Disease-free-interval (DFI) and overall survival (OS) were significantly associated with CST6 promoter methylation (p=0.004 and p=0.001 respectively). Multivariate analysis revealed that CST6 methylation is an independent prognostic factor for DFI (HR=3.484; 95% CI: 1.155-10.511; p=0.027). and OS (HR=9.190; 95% CI: 1.989-42.454; p=0.004). CST6 promoter methylation status in tumor cells seems to provide important prognostic information in operable breast cancer and merits to be further evaluated and validated in a larger cohort of patients.
Collapse
Affiliation(s)
- Magdalini Kioulafa
- Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
153
|
Yoon NK, Seligson DB, Chia D, Elshimali Y, Sulur G, Li A, Horvath S, Maresh E, Mah V, Bose S, Bonavida B, Goodglick L. Higher expression levels of 14-3-3sigma in ductal carcinoma in situ of the breast predict poorer outcome. Cancer Biomark 2009; 5:215-24. [PMID: 19729831 DOI: 10.3233/cbm-2009-0106] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The protein 14-3-3sigma is involved in the regulation of cellular processes such as apoptosis, cell cycle progression and proliferation. Disruption of protein expression has been implicated in a number of malignancies. Here we examine the expression pattern of 14-3-3sigma in breast cancer and specifically consider whether expression in ductal carcinoma in situ (DCIS) lesions is predictive of disease outcome. We examined 14-3-3sigma protein expression and localization using immunohistochemical staining on a high-density tissue microarray consisting of 157 invasive breast cancer patients. Statistical analyses were used to assess the correlation of 14-3-3sigma expression with clinico-pathological parameters and patient outcome. We observed a statistically significant increase in 14-3-3sigma protein expression in ductal hyperplasia, DCIS, and invasive ductal carcinoma (IDC) as compared normal glandular epithelium. In IDC, lower expression of 14-3-3sigma tended to predicted poorer survival time while in DCIS lesions, there was a stronger correlation between relatively higher levels of 14-3-3sigma predicting shorter survival time. Further, of patients who had concurrent DCIS and IDC lesions, those that exhibited a decrease of 14-3-3sigma expression from DCIS to IDC had significantly shorter survival time. Our findings indicate that 14-3-3sigma expression may be a useful prognostic indicator for survival in patients with breast cancer with an elevated 14-3-3sigma in earlier disease predicting a less favorable disease outcome. To our knowledge this is the first published study associating 14-3-3sigma protein expression with breast cancer survival.
Collapse
Affiliation(s)
- Nam K Yoon
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1747, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Genomic screening for genes upregulated by demethylation revealed novel targets of epigenetic silencing in breast cancer. Breast Cancer Res Treat 2009; 122:699-710. [PMID: 19859801 DOI: 10.1007/s10549-009-0600-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 10/12/2009] [Indexed: 01/14/2023]
Abstract
Breast cancer arises through the accumulation of multiple genetic alterations and epigenetic changes such as methylation, which silences gene expression in a variety of cancers. In the present study, we applied genomic screening to identify genes upregulated by the demethylating agent 5-aza-2'-deoxycytidine (DAC) in a human breast cancer cell line (MCF7). We identified 288 genes upregulated and 29 genes downregulated more than fivefold after treatment with DAC, and gene ontology analyses revealed the genes to be involved in immune responses, apoptosis, and cell differentiation. In addition, real-time PCR analysis of ten genes silenced in MCF7 cells confirmed that they are upregulated by DAC, while bisulfite-pyrosequencing analysis confirmed that nine of those genes were silenced by methylation. We also found that treating MCF7 cells with DAC restored induction of DFNA5 by p53, as well as by two other p53 family genes, p63gamma and p73beta. Introduction of NTN4 into MCF7 cells suppressed cell growth, indicating that NTN4 has tumor suppressive activity. In primary breast cancers, we detected cancer-specific methylation of NTN4, PGP9.5, and DKK3, suggesting that methylation of these genes could be useful markers for diagnosis of breast cancer. Thus, DNA methylation appears to be a common event in breast cancer, and the genes silenced by methylation could be useful targets for both diagnosis and therapy.
Collapse
|
155
|
Translational control during early development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 90:211-54. [PMID: 20374743 DOI: 10.1016/s1877-1173(09)90006-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Translational control of specific messenger RNAs, which themselves are often asymmetrically localized within the cytoplasm of a cell, underlies many events in germline development, and in embryonic axis specification. This comprehensive, but by no means exhaustive, review attempts to present a picture of the present state of knowledge about mechanisms underlying mRNA localization and translational control of specific mRNAs that are mediated by trans-acting protein factors. While RNA localization and translational control are widespread in evolution and have been studied in many experimental systems, this article will focus mainly on three particularly well-characterized systems: Drosophila, Caenorhabditis elegans, and Xenopus. In keeping with the overall theme of this volume, instances in which translational control factors have been linked to human disease states will also be discussed.
Collapse
|
156
|
Hayashi E, Kuramitsu Y, Fujimoto M, Zhang X, Tanaka T, Uchida K, Fukuda T, Furumoto H, Ueyama Y, Nakamura K. Proteomic profiling of differential display analysis for human oral squamous cell carcinoma: 14-3-3 σ Protein is upregulated in human oral squamous cell carcinoma and dependent on the differentiation level. Proteomics Clin Appl 2009; 3:1338-47. [PMID: 21136954 DOI: 10.1002/prca.200900091] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 08/05/2009] [Accepted: 08/08/2009] [Indexed: 01/08/2023]
Abstract
Oral squamous cell carcinoma (OSCC) has an absolute majority of all oral cancer. We used proteomic technology to analyze the protein expression profile in OSCC tissues and accompanying surrounding normal tissues in four oral locations (buccal mucosa, gingival mucosa, oral floor, and tongue). Ten protein spots were overexpressed more strongly in cancer tissues than normal ones, and were identified as proliferating cell nuclear antigen, 14-3-3 ε, 14-3-3 σ, proteasome subunit α type 5, translationally controlled tumor protein, eukaryotic translation initiation factor 3 subunit, macrophage capping protein, and mitochondrial isocitrate dehydrogenase subunit α. Macrophage capping protein and mitochondrial isocitrate dehydrogenase subunit α had two spots. Especially, we focused on 14-3-3 σ protein, one of the eight identified proteins, and assessed its expression level in four oral locations of OSCC by using differential display methods. The expression level of 14-3-3 σ protein was upregulated in four locations of oral cavity. Eight proteins which we identified in this study may play an important role in OSCC carcinogenesis and progression and could be used as diagnostic biomarkers of OSCC.
Collapse
Affiliation(s)
- Eiko Hayashi
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan; Department of Oral and Maxillofacial Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Zhang M, Yang H. Negative growth regulators of the cell cycle machinery and cancer. PATHOPHYSIOLOGY 2009; 16:305-9. [DOI: 10.1016/j.pathophys.2009.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
158
|
Aberrant methylation profile of 14-3-3 sigma and its reduced transcription/expression levels in Chinese sporadic female breast carcinogenesis. Med Oncol 2009; 27:791-7. [PMID: 19685192 DOI: 10.1007/s12032-009-9287-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 08/02/2009] [Indexed: 12/21/2022]
Abstract
To study the relation of 14-3-3 sigma gene promoter hypermethylation and its transcription expression levels in sporadic breast carcinogenesis. Methylation of 14-3-3 sigma gene was detected by sensitive MSP assay in carcinous, non-cancerous, and normal tissue, and its mRNA was also detected by real time PCR based on SYBR Green 1 as well, and protein was detected by west blotting assay. The methylation frequencies of 14-3-3 sigma were 90% in 68 cases of sporadic breast cancer patients. Methylation was presented in portions (2/13, 18%) of hyperplastic samples, and no hypermethylation was presented in normal tissue. The methylation change of 14-3-3 sigma gene was markedly related with various types, grades, and lymph node metastases (P < 0.05), and no significant differences in methylation frequencies were seen between premenopause and postmenopause (P > 0.05). The methylation of 14-3-3 sigma shows reverse relation with its mRNA transcription and expression level (P < 0.05). Only was lymph node metastases strongly associated with poor outcome (P = 0.02). Whether 14-3-3 sigma promoter methylation or not did not affect the 5 years survival rate of sporadic breast cancer patients (P > 0.05). Epigenetics alterations of the 14-3-3 sigma can contribute to reducing or losing expression of 14-3-3 sigma protein, which play an important role in the development of sporadic breast carcinomas and involved in various types, grades, and lymph node metastases. Otherwise, node metastases of breast carcinogenesis patients are strongly associated with poor outcome.
Collapse
|
159
|
Wang X, Chen Y, Han QB, Chan CY, Wang H, Liu Z, Cheng CHK, Yew DT, Lin MCM, He ML, Xu HX, Sung JJY, Kung HF. Proteomic identification of molecular targets of gambogic acid: role of stathmin in hepatocellular carcinoma. Proteomics 2009; 9:242-53. [PMID: 19086098 DOI: 10.1002/pmic.200800155] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Gamboge has been developed as an injectable drug for cancer treatment in China. In this study, the inhibition ratio and their IC(50) values of two derivatives from Gamboge in hepatocellular carcinoma (HCC) were determined. Proteomic approach was employed to reveal the target proteins of these two derivatives, gambogic acid (GA), and gambogenic acid (GEA). HCC cells were cultured under varied conditions with the addition of either GA or GEA. Twenty differentially expressed proteins were identified and the four most distinctly expressed proteins were further validated by Western blotting. GA and GEA revealed inhibitory effects on HCC cell proliferation. The expression of cyclin-dependent kinase 4 inhibitor A and guanine nucleotide-binding protein beta subunit 1 were upregulated by both xanthones, whilst the expression of 14-3-3 protein sigma and stathmin 1 (STMN1) were downregulated. Furthermore, overexpression of STMN1 in HCC cells decreased their sensitivity, whilst small interfering RNAs targeting STMN1 enhanced their sensitivity to GA and GEA. In conclusion, our study suggested for the first time that STMN1 might be a major target for GA and GEA in combating HCC. Further investigation may lead to a new generation of anticancer drugs exerting synergistic effect with conventional therapy, thus to promote treatment efficacy.
Collapse
Affiliation(s)
- Xin Wang
- State Key Laboratory of Oncology in Southern China and Stanley Ho Center for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Schultz J, Ibrahim SM, Vera J, Kunz M. 14-3-3sigma gene silencing during melanoma progression and its role in cell cycle control and cellular senescence. Mol Cancer 2009; 8:53. [PMID: 19642975 PMCID: PMC2723074 DOI: 10.1186/1476-4598-8-53] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2009] [Accepted: 07/30/2009] [Indexed: 12/19/2022] Open
Abstract
Background The family of 14-3-3 proteins plays an important role in cancer biology by interfering with intracellular signalling pathways and cell cycle checkpoints. The 14-3-3σ isoform acts as a tumor suppressor and is often inactivated during tumor development. Results Here, we demonstrate enhanced CpG methylation of the 14-3-3σ gene in lymph node and cutaneous melanoma metastases compared with primary tumors, associated with dramatically reduced mRNA expression. In line with this, treatment of different metastatic melanoma cell lines with 5-aza-2'-deoxycytidine (5-Aza-CdR), a potent inhibitor of cytosine methylation, significantly induces 14-3-3σ protein expression. Additional treatment with histone deacetylase inhibitor 4-phenylbutyric acid (Pba) further enhances 14-3-3σ expression. Induction of 14-3-3σ expression by 5-Aza-CdR/Pba treatment leads to almost complete inhibition of cell proliferation, with cells predominantly arrested in G2-M. The antiproliferative effect of 5-Aza-CdR/Pba was reversed in 14-3-3σ knockdown cells. Similarly, melanoma cell lines stably overexpressing 14-3-3σ show dramatically reduced cell proliferation rates. Moreover, synchronous 14-3-3σ stably overexpressing cells do not progress through cell cycle, but display a permanent increase in the population of 4n DNA containing cells. Interestingly, overexpression of 14-3-3σ induces senescence of melanoma cells and is involved in melanoma cell senescence under genotoxic stress. Finally, 14-3-3σ knockdown supports migratory capacity of melanoma cells in vitro, while 14-3-3σ overexpression has opposing effects. Conclusion Taken together, the present report indicates that epigenetic silencing of 14-3-3σ might contribute to tumor progression in malignant melanoma via loss of cell cycle control, impaired cellular senescence program and support of migratory capacity.
Collapse
Affiliation(s)
- Julia Schultz
- Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| | | | | | | |
Collapse
|
161
|
Zanello SB, Nayak R, Zanello LP, Farthing-Nayak P. Identification and Distribution of 14.3.3σ (Stratifin) in the Human Cornea. Curr Eye Res 2009; 31:825-33. [PMID: 17050274 DOI: 10.1080/02713680600878816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We demonstrate for the first time the expression of 14.3.3sigma, an epithelial cell differentiation marker, in human corneal epithelium. 14.3.3sigma appeared at 30 kDa, pI 4-5, in 2D gels of corneal extracts. We found no significant differences in 14.3.3sigma levels between healthy corneas and corneas from keratoconus, corneal dystrophy, and corneal edema patients. 14.3.3sigma immunofluorescence was observed in the cytoplasm and nucleus of epithelial cells and colocalized with cyclin-B1. 14.3.3sigma was secreted by HCE-2 cells; HCE-2-conditioned medium induced matrix metalloproteinase-1 in cultured keratocytes. In summary, our work presents evidence of 14.3.3sigma expression in corneal epithelium and elaborates over its possible implications in corneal pathologic conditions.
Collapse
Affiliation(s)
- Susana B Zanello
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona, USA.
| | | | | | | |
Collapse
|
162
|
Yi B, Tan SX, Tang CE, Huang WG, Cheng AL, Li C, Zhang PF, Li MY, Li JL, Yi H, Peng F, Chen ZC, Xiao ZQ. Inactivation of 14-3-3 sigma by promoter methylation correlates with metastasis in nasopharyngeal carcinoma. J Cell Biochem 2009; 106:858-66. [PMID: 19160382 DOI: 10.1002/jcb.22051] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
14-3-3 sigma, the downstream target of p53, is a negative regulator of cell cycle G2-M phase checkpoint in response to DNA damage. Our previous comparative proteomics study showed that 14-3-3 sigma was downregulated or lost in nasopharyngeal carcinoma (NPC) tissue compared with non-cancerous nasopharyngeal epithelial tissue (NNET). In this study, we further investigated for the epigenetic mechanism of 14-3-3 sigma inactivation. Methylation-specific PCR showed 14-3-3 sigma promoter methylation in 100% of analyzed NPC cell lines (4/4) but not in immortalized human nasopharyngeal epithelial cell line NP69. Treatment of the four NPC cell lines with the methyltransferase inhibitor 5-aza-2'-dC resulted in the demethylation and upregulation of 14-3-3 sigma. In tissues, 14-3-3 sigma promoter methylation occurred at a higher frequency in NPC, 63/75 (84%), compared to adjacent NNET, 7/25 (28%), and fully methylated 14-3-3 sigma promoter was detected in NPC but not in any of adjacent NNET. RT-PCR, Western blotting, and immunohistochemistry showed that 14-3-3 sigma expression was downregulated or lost in NPC with methylation, and there was a negative correlation between the expression levels and methylation statuses of 14-3-3 sigma gene. In addition, the patients with methylated 14-3-3 sigma presented a higher frequency of lymph node and distant metastasis, and an advanced clinical stage, and overexpression of 14-3-3 sigma in NPC cell line 5-8F with high metastatic potential was able to inhibit its in vitro invasive ability. Our data are the first to show that 14-3-3 sigma is frequently inactivated by promoter methylation in NPC and this aberrant methylation correlates with lymph node and distant metastasis.
Collapse
Affiliation(s)
- Bin Yi
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Li Z, Liu JY, Zhang JT. 14-3-3sigma, the double-edged sword of human cancers. Am J Transl Res 2009; 1:326-340. [PMID: 19956445 PMCID: PMC2780041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 06/02/2009] [Indexed: 05/28/2023]
Abstract
14-3-3sigma is a member of a highly conserved family of 14-3-3 proteins that are present in all eukaryotic organisms. 14-3-3sigma has been considered as a tumor suppressor with reduced expression in some human cancers while its increased expression causes resistance to anticancer agents and radiation that cause DNA damages. The increased expression of 14-3-3sigma may also predict poor prognosis in some human cancers. Thus, 14-3-3sigma may play an important role as a double-edged sword in human cancers, which may attribute to its property as a molecular chaperone by binding to various protein ligands important to many cellular processes such as cell cycle checkpoint regulation and apoptosis in response to DNA damages. In this article, we will review recent studies and progresses in understanding 14-3-3sigma as a double-edged sword in human cancers.
Collapse
Affiliation(s)
- Zhaomin Li
- Department of Pharmacology and Toxicology and IU Simon Cancer Center, Indiana University School of Medicine Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
164
|
Li DJ, Deng G, Xiao ZQ, Yao HX, Li C, Peng F, Li MY, Zhang PF, Chen YH, Chen ZC. Identificating 14-3-3 sigma as a lymph node metastasis-related protein in human lung squamous carcinoma. Cancer Lett 2009; 279:65-73. [DOI: 10.1016/j.canlet.2009.01.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Revised: 12/09/2008] [Accepted: 01/18/2009] [Indexed: 10/21/2022]
|
165
|
Liu S, Howell P, Ren S, Fodstad O, Riker AI. The 14-3-3sigma gene promoter is methylated in both human melanocytes and melanoma. BMC Cancer 2009; 9:162. [PMID: 19473536 PMCID: PMC2695475 DOI: 10.1186/1471-2407-9-162] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 05/27/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent evidence demonstrates that 14-3-3sigma acts as a tumor suppressor gene inactivated by methylation of its 5' CpG islands in epithelial tumor cells, while remaining un-methylated in normal human epithelia. The methylation analysis of 14-3-3sigma has been largely overlooked in melanoma. METHODS The methylation status of 14-3-3sigma CpG island in melanocytes and melanoma cells was analyzed by methylation-specific sequencing (MSS) and quantitative methylation-specific PCR (Q-MSP). 14-3-3sigma mRNA and protein expression in cell lines was detected by real-time RT-PCR and western blot. Melanoma cells were also treated by 5-aza-2'-deoxycytidine (DAC), a demethylating agent, and/or histone deacetylase inhibitor, Trichostatin A (TSA), to evaluate their effects on 14-3-3sigma gene expression. RESULTS 14-3-3sigma is hypermethylated in both human melanocytes and most melanoma cells in a lineage-specific manner, resulting in the silencing of 14-3-3sigma gene expression and the active induction of 14-3-3sigma mRNA and protein expression following treatment with DAC. We also observed a synergistic effect upon gene expression when DAC was combined with TSA. The promoter methylation status of 14-3-3sigma was analyzed utilizing Q-MSP in 20 melanoma tissue samples and 10 cell lines derived from these samples, showing that the majority of melanoma samples maintain their hypermethylation status of the 14-3-3sigma gene. CONCLUSION 14-3-3sigma is hypermethylated in human melanoma in a cell-linage specific manner. Spontaneous demethylation and re-expression of 14-3-3sigma is a rare event in melanoma, indicating 14-3-3sigma might have a tentative role in the pathogenesis of melanoma.
Collapse
Affiliation(s)
- Suhu Liu
- Ochsner Cancer Institute, Department of Surgery 1514 Jefferson Highway, BH334 New Orleans, LA 70121, USA.
| | | | | | | | | |
Collapse
|
166
|
Toyota M, Suzuki H, Yamashita T, Hirata K, Imai K, Tokino T, Shinomura Y. Cancer epigenomics: implications of DNA methylation in personalized cancer therapy. Cancer Sci 2009; 100:787-91. [PMID: 19236379 PMCID: PMC11159488 DOI: 10.1111/j.1349-7006.2009.01095.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Genetic alterations in cancer can provide information for predicting a tumor's sensitivity to chemotherapeutic drugs. But although such information is certainly useful, the relatively low frequency of mutations seen in many cancers limits the utility of pharmacogenomics in large numbers of cancer patients, necessitating consideration of other approaches. Epigenetic changes such as DNA methylation are a hallmark of human cancers. Methylation of genes involved in DNA repair and maintaining genome integrity (e.g. MGMT, hMLH1, WRN, and FANCF), and cell-cycle checkpoint genes (e.g. CHFR and 14-3-3 sigma, CDK10, and p73), all reportedly influence the sensitivity to chemotherapeutic drugs, suggesting that DNA methylation could serve as a molecular marker for predicting the responsiveness of tumors to chemotherapy. However, the comprehensive study of pharmacoepigenomics awaits the advent of genome-wide analysis of DNA methylation using microarrays and next-generation sequencers.
Collapse
Affiliation(s)
- Minoru Toyota
- Department of Biochemistry, Cancer Research Institute, Sapporo Medical University, Sapporo 060-8556, Japan.
| | | | | | | | | | | | | |
Collapse
|
167
|
Neal CL, Yao J, Yang W, Zhou X, Nguyen NT, Lu J, Danes CG, Guo H, Lan KH, Ensor J, Hittelman W, Hung MC, Yu D. 14-3-3zeta overexpression defines high risk for breast cancer recurrence and promotes cancer cell survival. Cancer Res 2009; 69:3425-32. [PMID: 19318578 PMCID: PMC2671640 DOI: 10.1158/0008-5472.can-08-2765] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The ubiquitously expressed 14-3-3 proteins are involved in numerous important cellular functions. The loss of 14-3-3sigma is a common event in breast cancer; however, the role of other 14-3-3s in breast cancer is unclear. Recently, we found that 14-3-3zeta overexpression occurs in early stage breast diseases and contributes to transformation of human mammary epithelial cells. Here, we show that 14-3-3zeta overexpression also persisted in invasive ductal carcinoma and contributed to the further progression of breast cancer. To examine the clinical effect of 14-3-3zeta overexpression in advanced stage breast cancer, we performed immunohistochemical analysis of 14-3-3zeta expression in primary breast carcinomas. 14-3-3zeta overexpression occurred in 42% of breast tumors and was determined to be an independent prognostic factor for reduced disease-free survival. 14-3-3zeta overexpression combined with ErbB2 overexpression and positive lymph node status identified a subgroup of patients at high risk for developing distant metastasis. To investigate whether 14-3-3zeta overexpression causally promotes breast cancer progression, we overexpressed 14-3-3zeta by stable transfection or reduced 14-3-3zeta expression by siRNA in cancer cell lines. Increased 14-3-3zeta expression enhanced anchorage-independent growth and inhibited stress-induced apoptosis, whereas down-regulation of 14-3-3zeta reduced anchorage-independent growth and sensitized cells to stress-induced apoptosis via the mitochondrial apoptotic pathway. Transient blockade of 14-3-3zeta expression by siRNA in cancer cells effectively reduced the onset and growth of tumor xenografts in vivo. Therefore, 14-3-3zeta overexpression is a novel molecular marker for disease recurrence in breast cancer patients and may serve as an effective therapeutic target in patients whose tumors overexpress 14-3-3zeta.
Collapse
Affiliation(s)
- Christopher L. Neal
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Jun Yao
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Wentao Yang
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Xiaoyan Zhou
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Nina T. Nguyen
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Jing Lu
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Christopher G. Danes
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Hua Guo
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Keng-Hsueh Lan
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Joe Ensor
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Walter Hittelman
- Department of Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
168
|
Zhu F, Park E, Liu B, Xia X, Fischer SM, Hu Y. Critical role of IkappaB kinase alpha in embryonic skin development and skin carcinogenesis. Histol Histopathol 2009; 24:265-71. [PMID: 19085841 PMCID: PMC7243875 DOI: 10.14670/hh-24.265] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
IkappaB kinase alpha (IKKalpha), IKKbeta, and IKKgamma/NEMO form the IKK complex, which is essential for NF-kappaB activation. However, genetic studies have shown that the role of IKKalpha is distinct from that of IKKbeta or IKKgamma in the development of the mouse embryonic skin. Loss of IKKalpha has been shown to cause epidermal hyperplasia, prevent keratinocyte terminal differentiation, and impair the formation of the skin, resulting in the deaths of IKKalpha-deficient (Ikkalpha-/-) mice soon after birth. Recent experimental data from several laboratories have revealed that IKKalpha functions as a tumor suppressor in human squamous cell carcinomas (SCCs) of skin, lungs, and head and neck. Chemical carcinogenesis studies using mice have shown that reduction in IKKalpha expression increases the number and size of Ras-initiated skin tumors and promotes their progression, indicating that reduced IKKalpha expression provides a selective growth advantage that cooperates with Ras activity to promote skin carcinogenesis. In this review, we will summarize these findings from our and other studies on the role that IKKalpha plays in development of the mouse embryonic skin and skin carcinogenesis.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Eunmi Park
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Bigang Liu
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Xiaojun Xia
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Susan M. Fischer
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Yinling Hu
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
- Cancer and Inflammation Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, USA
| |
Collapse
|
169
|
Hütter G, Letsch A, Nowak D, Poland J, Sinha P, Thiel E, Hofmann WK. High correlation of the proteome patterns in bone marrow and peripheral blood blast cells in patients with acute myeloid leukemia. J Transl Med 2009; 7:7. [PMID: 19146667 PMCID: PMC2630935 DOI: 10.1186/1479-5876-7-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 01/15/2009] [Indexed: 11/13/2022] Open
Abstract
Background When comparing myelogenous blasts from bone marrow and peripheral blood, immunophenotyping usually show a strong correlation of expression of surface antigens. However, it remains to be determined, whether this correlation also exists on the level of protein expression. Method Therefore, we investigated both bone marrow and peripheral blood blast cells from six patients with newly diagnosed acute myeloid leukemia (AML) using conventional two-dimensional electrophoresis in the first dimension and linear polyacrylamide gels (12%) in the second dimension. Proteins were visualized using the silver staining method and image analysis was performed using the PDQuest system. Results For each patient over 80 proteins were evaluated in the sample from peripheral blood and bone marrow. We could demonstrate that the protein expression profile of bone marrow did not significantly differ from the expression patterns of peripheral blast cells. Conclusion The proteome-set of leukemic blast cells from marrow and blood, does not differ substantially when drawn from AML patients with over 80 percent blast cells in both compartments. This indicates that in AML, blasts from peripheral blood samples can be considered suitable for investigations of the proteome using 2D-electrophoresis.
Collapse
Affiliation(s)
- Gero Hütter
- Department of Internal Medicine III (Hematology, Onkology), Charité Berlin Campus Benjamin Franklin, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
170
|
Stenzinger A, Schreiner D, Koch P, Hofer HW, Wimmer M. Cell and molecular biology of the novel protein tyrosine-phosphatase-interacting protein 51. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 275:183-246. [PMID: 19491056 DOI: 10.1016/s1937-6448(09)75006-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This chapter examines the current state of knowledge about the expression profile, as well as biochemical properties and biological functions of the evolutionarily conserved protein PTPIP51. PTPIP51 is apparently expressed in splice variants and shows a particularly high expression in epithelia, skeletal muscle, placenta, and germ cells, as well as during mammalian development and in cancer. PTPIP51 is an in vitro substrate of Src- and protein kinase A, the PTP1B/TCPTP protein tyrosine phosphatases and interacts with 14-3-3 proteins, the Nuf2 kinetochore protein, the ninein-interacting CGI-99 protein, diacylglycerol kinase alpha, and also with itself forming dimers and trimers. Although the precise cellular function remains to be elucidated, the current data implicate PTPIP51 in signaling cascades mediating proliferation, differentiation, apoptosis, and motility.
Collapse
Affiliation(s)
- Albrecht Stenzinger
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | |
Collapse
|
171
|
Demircan B, Dyer LM, Gerace M, Lobenhofer EK, Robertson KD, Brown KD. Comparative epigenomics of human and mouse mammary tumors. Genes Chromosomes Cancer 2009; 48:83-97. [PMID: 18836996 PMCID: PMC2929596 DOI: 10.1002/gcc.20620] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Gene silencing by aberrant epigenetic chromatin alteration is a well-recognized event contributing to tumorigenesis. Although genetically engineered tumor-prone mouse models have proven a powerful tool in understanding many aspects of carcinogenesis, to date few studies have focused on epigenetic alterations in mouse tumors. To uncover epigenetically silenced tumor suppressor genes (TSGs) in mouse mammary tumor cells, we conducted initial genome-wide screening by combining the treatment of cultured cells with the DNA demethylating drug 5-aza-2'-deoxycytidine (5-azadC) and the histone deacetylase inhibitor trichostatin A (TSA) with expression microarray. By conducting this initial screen on EMT6 cells and applying protein function and genomic structure criteria to genes identified as upregulated in response to 5-azadC/TSA, we were able to identify two characterized breast cancer TSGs (Timp3 and Rprm) and four putative TSGs (Atp1B2, Dusp2, FoxJ1 and Smpd3) silenced in this line. By testing a panel of 10 mouse mammary tumor lines, we determined that each of these genes is commonly hypermethylated, albeit with varying frequency. Furthermore, by examining a panel of human breast tumor lines and primary tumors we observed that the human orthologs of ATP1B2, FOXJ1 and SMPD3 are aberrantly hypermethylated in the human disease whereas DUSP2 was not hypermethylated in primary breast tumors. Finally, we examined hypermethylation of several genes targeted for epigenetic silencing in human breast tumors in our panel of 10 mouse mammary tumor lines. We observed that the orthologs of Cdh1, RarB, Gstp1, RassF1 genes were hypermethylated, whereas neither Dapk1 nor Wif1 were aberrantly methylated in this panel of mouse tumor lines. From this study, we conclude that there is significant, but not absolute, overlap in the epigenome of human and mouse mammary tumors.
Collapse
Affiliation(s)
- Berna Demircan
- Department of Biochemistry and Molecular Biology, UF-Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | |
Collapse
|
172
|
Vera J, Schultz J, Ibrahim S, Raatz Y, Wolkenhauer O, Kunz M. Dynamical effects of epigenetic silencing of 14-3-3σ expression. ACTA ACUST UNITED AC 2009; 6:264-73. [DOI: 10.1039/b907863k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
173
|
Abstract
Breast carcinogenesis involves genetic and epigenetic alterations that cause aberrant gene function. Recent progress in the knowledge of epigenomics has had a profound impact on the understanding of mechanisms leading to breast cancer, and consequently the development of new strategies for diagnosis and treatment of breast cancer. Epigenetic regulation has been known to involve three mutually interacting events--DNA methylation, histone modifications and nucleosomal remodeling. These processes modulate chromatin structure to form euchromatin or heterochromatin, and in turn activate or silence gene expression. Alteration in expression of key genes through aberrant epigenetic regulation in breast cells can lead to initiation, promotion and maintenance of carcinogenesis, and is even implicated in the generation of drug resistance. We currently review known roles of the epigenetic machinery in the development and recurrence of breast cancer. Furthermore, we highlight the significance of epigenetic alterations as predictive biomarkers and as new targets of anticancer therapy.
Collapse
Affiliation(s)
- Pang-Kuo Lo
- Johns Hopkins University School of Medicine, 1650 Orleans Street, CRBI-143, Baltimore, MD 21231, USA; Tel.: +1 410 614 2479 Fax: +1 410 614 4073
| | - Saraswati Sukumar
- Johns Hopkins University School of Medicine, 1650 Orleans Street, CRBI-143, Baltimore, MD 21231, USA; Tel.: +1 410 614 2479 Fax: +1 410 614 4073
- Breast Cancer Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, USA
| |
Collapse
|
174
|
Abstract
PURPOSE The purpose of the present study was to investigate the potential role of 14-3-3sigma in pancreatic ductal adenocarcinoma (PDAC). EXPERIMENTAL DESIGN 14-3-3 isoform expression was determined by real-time quantitative PCR in laser capture normal pancreatic ductal cells and pancreatic cancer cells and in 5 pancreatic cancer cell lines. PANC-1 cells, with low levels of 14-3-3sigma, were stably transfected with a human 14-3-3sigma cDNA. Conversely, high endogenous 14-3-3sigma levels in T3M4 cells were suppressed by specific short hairpin RNA. Apoptosis, motility, and invasiveness were studied. RESULTS The cancer cells in 7 PDAC samples expressed high levels of 14-3-3sigma mRNA by quantitative PCR when compared with normal pancreatic duct cells. 14-3-3sigma protein levels were high in BxPC3, COLO-357, and T3M4 cells, intermediate in ASPC-1 cells, and low in PANC-1 cells. Most cell lines released detectable amount of 14-3-3sigma into conditioned medium. Overexpression of 14-3-3sigma in PANC-1 cells led to resistance to cisplatinum-induced apoptosis, increased basal migration, and increased invasion in response to epidermal growth factor and insulin-like growth factor-I. By contrast, short hairpin RNA-mediated knockdown of endogenous 14-3-3sigma in T3M4 cells did not alter migration but led to enhanced cisplatinum sensitivity, increased invasiveness in response to epidermal growth factor, and decreased invasiveness in response to insulin-like growth factor-I. CONCLUSIONS 14-3-3sigma contributes to the chemoresistance of pancreatic cancer cells and exerts cell type-dependent effects on cell migration and invasion. Therefore, strategies aimed at suppressing 14-3-3sigma expression and function may have a therapeutic benefit in subgroups of patients with PDAC.
Collapse
Affiliation(s)
- Divas Neupane
- Department of Medicine and Pharmacology and Toxicology, Dartmouth-Hitchcock Medical Center and Dartmouth Medical School, Hanover, New Hamsphire 03756, USA
| | | |
Collapse
|
175
|
Suzuki H, Tokino T, Shinomura Y, Imai K, Toyota M. DNA methylation and cancer pathways in gastrointestinal tumors. Pharmacogenomics 2008; 9:1917-28. [DOI: 10.2217/14622416.9.12.1917] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer is fundamentally a genetic and epigenetic disease that requires the accumulation of genomic alterations that inactivate tumor suppressors and activate proto-oncogenes. In addition to genetic mutation or allelic loss, epigenetic gene silencing associated with DNA methylation is now recognized as an alternative mechanism by which tumor suppressor genes are inactivated. In gastrointestinal cancers, for example, DNA methylation frequently alters the activity in a number of important signaling pathways by silencing expression of genes encoding Wnt antagonists, negative Ras effectors and p53 targets. Indeed, the list of genes aberrantly methylated in cancer is growing, and methylation of a p53 target micoRNA gene has recently been demonstrated. Sites of DNA methylation could be promising markers and targets for risk assessment, early detection and treatment of cancer.
Collapse
Affiliation(s)
- Hiromu Suzuki
- First department of Internal Medicine, Sapporo Medical University, Japan
- Department of Biochemistry, Sapporo Medical University, S1, W17, Chuo-Ku, Sapporo, 060–8556, Japan
| | - Takashi Tokino
- Department of Molecular Biology, Cancer Research Institute, Sapporo Medical University, Japan
| | - Yasuhisa Shinomura
- First department of Internal Medicine, Sapporo Medical University, Japan
| | | | - Minoru Toyota
- Department of Biochemistry, Sapporo Medical University, S1, W17, Chuo-Ku, Sapporo, 060–8556, Japan
| |
Collapse
|
176
|
Kovacheva VP, Davison JM, Mellott TJ, Rogers AE, Yang S, O'Brien MJ, Blusztajn JK. Raising gestational choline intake alters gene expression in DMBA-evoked mammary tumors and prolongs survival. FASEB J 2008; 23:1054-63. [PMID: 19047067 DOI: 10.1096/fj.08-122168] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Choline is an essential nutrient that serves as a donor of metabolic methyl groups used during gestation to establish the epigenetic DNA methylation patterns that modulate tissue-specific gene expression. Because the mammary gland begins its development prenatally, we hypothesized that choline availability in utero may affect the gland's susceptibility to cancer. During gestational days 11-17, pregnant rats were fed a control, choline-supplemented, or choline-deficient diet (8, 36, and 0 mmol/kg of choline, respectively). On postnatal day 65, the female offspring received 25 mg/kg of a carcinogen 7,12-dimethylbenz[alpha]anthracene. Approximately 70% of the rats developed mammary adenocarcinomas; prenatal diet did not affect tumor latency, incidence, size, and multiplicity. Tumor growth rate was inversely related to choline content in the prenatal diet, resulting in 50% longer survival until euthanasia, determined by tumor size, of the prenatally choline-supplemented rats compared with the prenatally choline-deficient rats. This was accompanied by distinct expression patterns of approximately 70 genes in tumors derived from the three dietary groups. Tumors from the prenatally choline-supplemented rats overexpressed genes that confer favorable prognosis in human cancers (Klf6, Klf9, Nid2, Ntn4, Per1, and Txnip) and underexpressed those associated with aggressive disease (Bcar3, Cldn12, Csf1, Jag1, Lgals3, Lypd3, Nme1, Ptges2, Ptgs1, and Smarcb1). DNA methylation within the tumor suppressor gene, stratifin (Sfn, 14-3-3sigma), was proportional to the prenatal choline supply and correlated inversely with the expression of its mRNA and protein in tumors, suggesting that an epigenetic mechanism may underlie the altered molecular phenotype and tumor growth. Our results suggest a role for adequate maternal choline nutrition during pregnancy in prevention/alleviation of breast cancer in daughters.
Collapse
Affiliation(s)
- Vesela P Kovacheva
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | | | |
Collapse
|
177
|
Wang Z, Tropè CG, Suo Z, Trøen G, Yang G, Nesland JM, Holm R. The clinicopathological and prognostic impact of 14-3-3 sigma expression on vulvar squamous cell carcinomas. BMC Cancer 2008; 8:308. [PMID: 18950492 PMCID: PMC2577690 DOI: 10.1186/1471-2407-8-308] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Accepted: 10/24/2008] [Indexed: 11/13/2022] Open
Abstract
Background 14-3-3 sigma (σ) promotes G2/M cell cycle arrest by sequestering cyclin B1-CDC2 complex in cytoplasm. Down-regulation of 14-3-3σ, which has been demonstrated in various carcinomas, may contribute to malignant transformation. However, the exact role of 14-3-3σ in the pathogenesis of vulvar carcinoma is not fully characterized, and the prognostic impact of 14-3-3σ protein expression is still unknown. Methods We investigated the 14-3-3σ expression in a series of 302 vulvar squamous cell carcinomas using immunohistochemistry and its associations with clinicopathological factors and clinical outcome. Results In cytoplasm, nucleus and cytoplasm/nucleus of vulvar carcinomas high 14-3-3σ protein expression was found in 72%, 59% and 75% of the carcinomas, respectively, and low levels in 28%, 41% and 25% of the cases, respectively. High level of 14-3-3σ in cytoplasm, nucleus and cytoplasm/nucleus was significantly correlated to large tumor diameter (p = 0.001, p = 0.002 and p = 0.001, respectively) and deep invasion (p = 0.01, p = 0.001 and p = 0.007, respectively). Variations of 14-3-3σ protein expression were not associated to disease-specific survival. Conclusion Our results indicate that 14-3-3σ may be involved in the development of a subset of vulvar squamous cell carcinomas by down-regulation of 14-3-3σ protein. Neither cytoplasmic nor nuclear level of 14-3-3σ expression was associated with prognosis.
Collapse
Affiliation(s)
- Zhihui Wang
- Division of Pathology, The Norwegian Radium Hospital, Rikshospitalet University Hospital, Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
178
|
Liang S, Xu Y, Shen G, Zhao X, Zhou J, Li X, Gong F, Ling B, Fang L, Huang C, Wei Y. Gene expression and methylation status of 14-3-3sigma in human renal carcinoma tissues. IUBMB Life 2008; 60:534-40. [PMID: 18548556 DOI: 10.1002/iub.75] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Loss of 14-3-3sigma expression mainly by methylation-mediated silencing has been reported in several human cancers, but the methylation status of 14-3-3sigma in human renal carcinoma is rarely studied so far. In this report, 14-3-3sigma expression was first examined by RT-PCR and immunohistochemistry, and further we investigated the methylation status by methylation-specific PCR and the correlation between 14-3-3sigma expression and its methylation. We found 14-3-3sigma expression was lost in 27 of 31 renal tissues including 16 renal carcinoma tissues, eight para-cancerous kidney tissues and seven normal kidney tissues. Among 16 renal carcinoma tissues, 14 cases had complete hypermethylation of 14-3-3sigma. Eight para-cancerous kidney tissues were almost completely methylated except one case had both methylation and unmethylation. Among seven normal kidney tissues, five cases had partial methylation, and the other two cases were completely methylated. In addition, 14-3-3sigma mRNA had weak expression in OS-RC-2 cells, but it increased with gradual demethylation after treatment by a demethylation agent, 5-aza-2'-deoxycytidine. In general, 14-3-3sigma mRNA was mostly unexpressed, and its DNA frequently hypermethylated within 14-3-3sigma coding region was closely associated with the gene silencing in cancerous and para-cancerous kidney tissues. 14-3-3sigma was also frequently methylated and almost silencing in normal kidney tissues. However, the methylation frequency was gradually reinforced with the extent of malignancy from normal to para-cancerous and cancerous kidney tissues.
Collapse
Affiliation(s)
- Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Hashimoto H, Sudo T, Mikami Y, Otani M, Takano M, Tsuda H, Itamochi H, Katabuchi H, Ito M, Nishimura R. Germ cell specific protein VASA is over-expressed in epithelial ovarian cancer and disrupts DNA damage-induced G2 checkpoint. Gynecol Oncol 2008; 111:312-9. [PMID: 18805576 DOI: 10.1016/j.ygyno.2008.08.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 08/10/2008] [Accepted: 08/12/2008] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Cancer cells have characteristics, such as high telomerase activity and high levels of migration activity and proliferation, which are very similar to those of germ cell lineages. In this study, we examined the expression of VASA, a germ cell lineage specific marker and evaluated its clinical significance in epithelial ovarian cancer (EOC). METHODS We investigated VASA expression in 75 EOC tissues by immunohistochemistry, correlating results with clinicopathological factors. To clarify the effects of VASA on cellular phenotypes, we compared the protein expression profiles between SKOV-3 cells stably expressing VASA (SKOV-3-VASA) and vector-control cell lines by coupling 2D fingerprinting and identification of proteins by mass spectrometry. RESULTS VASA expression in tumor cells was found in 21 of 75 cases and was positively correlated with high age and serous histology. Significant down-regulation of 14-3-3sigma was observed in SKOV-3-VASA versus control cells. Over-expression of VASA abrogates the G2 checkpoint, induced by DNA damage, by down-regulating the expression of 14-3-3sigma. CONCLUSIONS These results suggest that VASA may either play a direct role in the progression of EOC or serve as a valuable marker of tumorigenesis.
Collapse
|
180
|
Hinshelwood RA, Clark SJ. Breast cancer epigenetics: normal human mammary epithelial cells as a model system. J Mol Med (Berl) 2008; 86:1315-28. [PMID: 18716754 DOI: 10.1007/s00109-008-0386-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 06/17/2008] [Accepted: 06/25/2008] [Indexed: 12/23/2022]
Abstract
DNA hypermethylation and histone modifications are two critical players involved in epigenetic regulation and together play an important role in silencing tumor-suppressor genes in all cancers, including breast cancer. One of the major challenges facing breast cancer researchers is the problem of how to identify critical genes that are epigenetically silenced early in cancer initiation as these genes provide potential early diagnostic and/or therapeutic targets for breast cancer management. This review will focus on compelling evidence that normal Human Mammary Epithelial Cells (HMECs) that escape senescence in culture mimic genetic and epigenetic events occurring in early breast cancer, and provide a valuable system to delineate the early steps in epigenetic deregulation that often occur during transition of a normal breast cell to a premalignant cell. In particular, this model system has been used to investigate the relationship between gene silencing, DNA methylation, histone modifications, and polycomb association that may occur early in oncogenic transformation.
Collapse
Affiliation(s)
- Rebecca A Hinshelwood
- The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | | |
Collapse
|
181
|
Lal G, Padmanabha L, Provenzano M, Fitzgerald M, Weydert J, Domann FE. Regulation of 14-3-3sigma expression in human thyroid carcinoma is epigenetically regulated by aberrant cytosine methylation. Cancer Lett 2008; 267:165-74. [PMID: 18440129 PMCID: PMC2710378 DOI: 10.1016/j.canlet.2008.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 02/13/2008] [Accepted: 03/12/2008] [Indexed: 11/25/2022]
Abstract
Increased 14-3-3sigma expression has been observed by immunohistochemistry in papillary and anaplastic tumors, but not follicular thyroid cancers. 14-3-3sigma mRNA expression and methylation status was examined in tumor cell lines and primary thyroid tissues using real-time RT-PCR, bisulfite sequencing and methylation-specific PCR. Most of the 27 CpG's in the gene's CpG island were methylated in normal thyroid, TPC-1, NPA, FTC-238 and 2-7, which did not express 14-3-3sigma. In contrast, they were unmethylated in KAK-1 and anaplastic lines KAT4 and DRO-90. 14-3-3sigma expression was not increased in thyroid carcinomas, the majority of which had a methylated CpG island. In addition, 5-aza-dC treatment increased 14-3-3sigma expression in the FTC-238 and NPA cell lines, which had low baseline expression. We conclude 14-3-3sigma expression in thyroid carcinomas is regulated by CpG island hypermethylation.
Collapse
Affiliation(s)
- Geeta Lal
- Department of Surgery, Division of Surgical Oncology, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, 4641 JCP, Iowa city, IA 52242, USA.
| | | | | | | | | | | |
Collapse
|
182
|
Negraes PD, Favaro FP, Camargo JLV, Oliveira MLCS, Goldberg J, Rainho CA, Salvadori DMF. DNA methylation patterns in bladder cancer and washing cell sediments: a perspective for tumor recurrence detection. BMC Cancer 2008; 8:238. [PMID: 18702824 PMCID: PMC2527332 DOI: 10.1186/1471-2407-8-238] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 08/14/2008] [Indexed: 12/31/2022] Open
Abstract
Background Epigenetic alterations are a hallmark of human cancer. In this study, we aimed to investigate whether aberrant DNA methylation of cancer-associated genes is related to urinary bladder cancer recurrence. Methods A set of 4 genes, including CDH1 (E-cadherin), SFN (stratifin), RARB (retinoic acid receptor, beta) and RASSF1A (Ras association (RalGDS/AF-6) domain family 1), had their methylation patterns evaluated by MSP (Methylation-Specific Polymerase Chain Reaction) analysis in 49 fresh urinary bladder carcinoma tissues (including 14 cases paired with adjacent normal bladder epithelium, 3 squamous cell carcinomas and 2 adenocarcinomas) and 24 cell sediment samples from bladder washings of patients classified as cancer-free by cytological analysis (control group). A third set of samples included 39 archived tumor fragments and 23 matched washouts from 20 urinary bladder cancer patients in post-surgical monitoring. After genomic DNA isolation and sodium bisulfite modification, methylation patterns were determined and correlated with standard clinic-histopathological parameters. Results CDH1 and SFN genes were methylated at high frequencies in bladder cancer as well as in paired normal adjacent tissue and exfoliated cells from cancer-free patients. Although no statistically significant differences were found between RARB and RASSF1A methylation and the clinical and histopathological parameters in bladder cancer, a sensitivity of 95% and a specificity of 71% were observed for RARB methylation (Fisher's Exact test (p < 0.0001; OR = 48.89) and, 58% and 17% (p < 0.05; OR = 0.29) for RASSF1A gene, respectively, in relation to the control group. Conclusion Indistinct DNA hypermethylation of CDH1 and SFN genes between tumoral and normal urinary bladder samples suggests that these epigenetic features are not suitable biomarkers for urinary bladder cancer. However, RARB and RASSF1A gene methylation appears to be an initial event in urinary bladder carcinogenesis and should be considered as defining a panel of differentially methylated genes in this neoplasia in order to maximize the diagnostic coverage of epigenetic markers, especially in studies aiming at early recurrence detection.
Collapse
Affiliation(s)
- Priscilla D Negraes
- Department of Genetics, Biosciences Institute, UNESP, Sao Paulo State University, Botucatu, Sao Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
183
|
Laimer K, Blassnig N, Spizzo G, Kloss F, Rasse M, Obrist P, Schäfer G, Perathoner A, Margreiter R, Amberger A. Prognostic significance of 14-3-3sigma expression in oral squamous cell carcinoma (OSCC). Oral Oncol 2008; 45:127-34. [PMID: 18620895 DOI: 10.1016/j.oraloncology.2008.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 04/09/2008] [Accepted: 04/10/2008] [Indexed: 11/27/2022]
Abstract
14-3-3sigma an intracellular phosphoserine binding protein regulates different cellular signalling processes and is involved in cancer development. In this study, we examined the expression of 14-3-3sigma and evaluated its clinical significance in OSCC. Tumour tissue from 95 OSCC patients was analysed for 14-3-3sigma and p53 expression, respectively. The correlation of these proteins with survival and clinical parameters was assessed. 14-3-3sigma high expression was observed in 44.2% of OSCC patients. A significant role of 14-3-3sigma expression on survival was shown by Kaplan-Meier analysis. Median survival time was 4.1years for patients with 14-3-3sigma low tumours, compared with 1.36years for 14-3-3sigma high tumours (P=.0021). Subset analysis in patients receiving adjuvant chemotherapy showed that the overall survival was significantly decreased in 14-3-3sigma high tumours than in 14-3-3sigma low tumours (P=.02). p53 expression was not significant in univariate analyses. In multivariate regression analysis, 14-3-3sigma expression emerged as a significant independent parameter (P=.003). These results provide evidence that 14-3-3sigma expression is involved in OSCC and, in contrast to p53 expression represents a new prognostic marker for OSCC and therapy response. Pending validation targeting 14-3-3sigma might also be a new opportunity to improve therapy.
Collapse
Affiliation(s)
- Klaus Laimer
- Division of Maxillofacial Surgery, Innsbruck Medical University, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Moreira JMA, Shen T, Ohlsson G, Gromov P, Gromova I, Celis JE. A combined proteome and ultrastructural localization analysis of 14-3-3 proteins in transformed human amnion (AMA) cells: definition of a framework to study isoform-specific differences. Mol Cell Proteomics 2008; 7:1225-1240. [PMID: 18378962 DOI: 10.1074/mcp.m700439-mcp200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024] Open
Abstract
The 14-3-3 proteins constitute a family of highly conserved and broadly expressed multifunctional polypeptides that are involved in a variety of important cellular processes that include cell cycle progression, growth, differentiation, and apoptosis. Although the exact cellular function(s) of 14-3-3 proteins is not fully elucidated, as a rule these proteins act by binding to protein ligands, thus regulating their activity; so far more than 300 cellular proteins have been reported to interact with 14-3-3 proteins. Binding to cognate interacting partners is isoform-specific, but redundancy also exists as several binding peptides can be recognized by all isoforms, and some functions can be carried out by any isoform indistinctly. Moreover by interacting with different ligands in a spatially and temporally regulated fashion the same isoform can play multiple possibly even opposing roles where the resultant cellular outcome will be determined by the integration of the various effects. Although there is a large body of literature on specific aspects of 14-3-3 biology, not much is known on the coordinated aspects of 14-3-3 isoform expression, post-translational modifications, and subcellular localization. To address the question of isoform-specific differences, we carried out a comparative analysis of the patterns of expression, phosphorylation, and subcellular localization of the 14-3-3 beta, epsilon, sigma, tau, and zeta protein isoforms in transformed human amnion (AMA) cells. To validate as well as broaden our observations we analyzed the occurrence of the various isoforms in a large number of established cell lines and mammary and urothelial tissue specimens. Given the systematic approach we undertook and our application of isoform-discriminating technologies to the analysis of various cellular systems, we expect the data presented in this study to serve as an enabling resource for researchers working with 14-3-3 proteins.
Collapse
Affiliation(s)
- José M A Moreira
- Department of Proteomics in Cancer, Institute of Cancer Biology and Danish Centre for Translational Breast Cancer Research (DCTB), Danish Cancer Society, DK-2100 Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
185
|
Metge BJ, Frost AR, King JA, Dyess DL, Welch DR, Samant RS, Shevde LA. Epigenetic silencing contributes to the loss of BRMS1 expression in breast cancer. Clin Exp Metastasis 2008; 25:753-63. [PMID: 18566899 PMCID: PMC2763604 DOI: 10.1007/s10585-008-9187-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Accepted: 05/20/2008] [Indexed: 01/13/2023]
Abstract
Breast Cancer Metastasis Suppressor 1 (BRMS1) suppresses metastasis of human breast cancer, ovarian cancer and melanoma in athymic mice. Studies have also shown that BRMS1 is significantly downregulated in some breast tumors, especially in metastatic disease. However, the mechanisms which regulate BRMS1 expression are currently unknown. Upon examination of the BRMS1 promoter region by methylation specific PCR (MSP) analysis, we discovered a CpG island (-3477 to -2214), which was found to be hypermethylated across breast cancer cell lines. A panel of 20 patient samples analyzed showed that 45% of the primary tumors and 60% of the matched lymph node metastases, displayed hypermethylation of BRMS1 promoter. Furthermore, we found a direct correlation between the methylation status of the BRMS1 promoter in the DNA isolated from tissues, with the loss of BRMS1 expression assessed by immunohistochemistry. There are several studies investigating the mechanism by which BRMS1 suppresses metastasis; however thus far there is no study that reports the cause(s) of loss of BRMS1 expression in aggressive breast cancer. Here we report for the first time that BRMS1 is a novel target of epigenetic silencing; and aberrant methylation in the BRMS1 promoter may serve as a cause of loss of its expression.
Collapse
Affiliation(s)
- Brandon J. Metge
- Department of Oncologic Sciences, USA-Mitchell Cancer Institute, University of South Alabama, 307 N. University Blvd., Mobile, AL 36688, USA
| | - Andra R. Frost
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Judy A. King
- Department of Pharmacology, University of South Alabama, Mobile, AL, USA
- Department of Pathology, University of South Alabama, Mobile, AL, USA
| | - Donna Lynn Dyess
- Department of Surgery, University of South Alabama, Mobile, AL, USA
| | - Danny R. Welch
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajeev S. Samant
- Department of Oncologic Sciences, USA-Mitchell Cancer Institute, University of South Alabama, 307 N. University Blvd., Mobile, AL 36688, USA
| | - Lalita A. Shevde
- Department of Oncologic Sciences, USA-Mitchell Cancer Institute, University of South Alabama, 307 N. University Blvd., Mobile, AL 36688, USA
| |
Collapse
|
186
|
Agarwal M, Pandita S, Hunt CR, Gupta A, Yue X, Khan S, Pandita RK, Pratt D, Shay JW, Taylor JSA, Pandita TK. Inhibition of telomerase activity enhances hyperthermia-mediated radiosensitization. Cancer Res 2008; 68:3370-8. [PMID: 18451164 DOI: 10.1158/0008-5472.can-07-5831] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hyperthermia is a potent sensitizer of cell killing by ionizing radiation (IR); however, hyperthermia also induces heat shock protein 70 (HSP70) synthesis and HSP70 expression is associated with radioresistance. Because HSP70 interacts with the telomerase complex and expression of the telomerase catalytic unit (hTERT) extends the life span of the human cells, we determined if heat shock influences telomerase activity and whether telomerase inhibition enhances heat-mediated IR-induced cell killing. In the present study, we show that moderate hyperthermia (43 degrees C) enhances telomerase activity. Inhibition of telomerase activity with human telomerase RNA-targeted antisense agents, and in particular GRN163L, results in enhanced hyperthermia-mediated IR-induced cell killing, and ectopic expression of catalytic unit of telomerase (TERT) decreased hyperthermia-mediated IR-induced cell killing. The increased cell killing by heat and IR exposure in telomerase-inhibited cells correlates with delayed appearance and disappearance of gamma-H2AX foci as well as decreased chromosome repair. These results suggest that inactivation of telomerase before combined hyperthermia and radiotherapy could improve tumor killing.
Collapse
Affiliation(s)
- Manjula Agarwal
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63108, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Yang Y, Haitchi HM, Cakebread J, Sammut D, Harvey A, Powell RM, Holloway JW, Howarth P, Holgate ST, Davies DE. Epigenetic mechanisms silence a disintegrin and metalloprotease 33 expression in bronchial epithelial cells. J Allergy Clin Immunol 2008; 121:1393-9, 1399.e1-14. [PMID: 18423563 DOI: 10.1016/j.jaci.2008.02.031] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 02/25/2008] [Accepted: 02/26/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND A disintegrin and metalloprotease 33 (ADAM33) polymorphism is strongly associated with asthma and bronchial hyperresponsiveness. Although considered to be a mesenchymal cell-specific gene, recent reports have suggested epithelial expression of ADAM33 in patients with severe asthma. OBJECTIVES Because dysregulated expression of ADAM33 can contribute to disease pathogenesis, we characterized the mechanism or mechanisms that control its transcription and investigated ADAM33 expression in bronchial biopsy specimens and brushings from healthy and asthmatic subjects. METHODS The ADAM33 promoter and CpG island methylation were analyzed by using bioinformatics, luciferase reporters, and bisulfite sequencing of genomic DNA. Epithelial-mesenchymal transition was induced by using TGF-beta1. ADAM33 mRNA was scrutinized in bronchial biopsy specimens and brushings by using reverse transcriptase-quantitative polymerase chain reaction, melt-curve analysis, and direct sequencing. RESULTS The predicted ADAM33 promoter (-550 to +87) had promoter transcriptional activity. Bisulfite sequencing showed that the predicted promoter CpG island (-362 to +80) was hypermethylated in epithelial cells but hypomethylated in ADAM33-expressing fibroblasts. Treatment of epithelial cells with 5-aza-deoxycytidine caused demethylation of the CpG island and induced ADAM33 expression. In contrast, phenotypic transformation of epithelial cells through a TGF-beta-induced epithelial-mesenchymal transition was insufficient to induce ADAM33 expression. ADAM33 mRNA was confirmed in bronchial biopsy specimens, but no validated signal was detected in bronchial brushings from healthy or asthmatic subjects. CONCLUSION The ADAM33 gene contains a regulatory CpG island within its promoter, the methylation status of which tightly controls its expression in a cell type-specific manner. ADAM33 repression is a stable feature of airway epithelial cells, irrespective of disease.
Collapse
Affiliation(s)
- Youwen Yang
- Brooke Laboratories, Division of Infection, Inflammation and Repair, School of Medicine, University of Southampton, Southampton, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Feng W, Shen L, Wen S, Rosen DG, Jelinek J, Hu X, Huan S, Huang M, Liu J, Sahin AA, Hunt KK, Bast RC, Shen Y, Issa JPJ, Yu Y. Correlation between CpG methylation profiles and hormone receptor status in breast cancers. Breast Cancer Res 2008; 9:R57. [PMID: 17764565 PMCID: PMC2206733 DOI: 10.1186/bcr1762] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Revised: 07/13/2007] [Accepted: 08/31/2007] [Indexed: 12/23/2022] Open
Abstract
Introduction Aberrant DNA methylation has been found frequently in human breast cancers, associated with the loss of expression of a number of regulatory genes for growth and correlated to clinical outcomes. The present study was undertaken to determine whether methylation of a set of growth-suppressor genes would correlate to the expression of estrogen receptors (ERs) and progesterone receptors (PRs). Methods We used a pyrosequencing methylation analysis to study the methylation of 12 known growth-suppressor genes in 90 pairs of malignant/normal breast tissues. We also examined the expression of ERs and PRs in those specimens by immunohistochemistry. Mutations of p53 in tumor cells were detected by direct sequencing. Results Twelve tumor-suppressor genes: ARHI, RASSF1A, HIN-1, RARβ2, hMLH1, 14-3-3 σ, RIZ1, p16, E-cadherin, RIL, CDH13, and NKD2 were selected for this methylation study. Five of them (RIL, HIN-1, RASSF1A, CDH13, and RARβ2) were frequently methylated in breast cancers (57%, 49%, 58%, 44%, and 17%, respectively) but not the normal breast (0–4%). Two panels of methylation profiles were defined. The methylation of the HIN-1/RASSFIA panel strongly correlated to the expression of ERs, PRs, and hormone receptors (HRs; which were defined as 'positive' if ERs and/or PRs were positive; p < 0.001). Conversely, the methylation of the RIL/CDH13 panel strongly correlated to negative ER, PR, and HR expression (p = 0.001, 0.025, and 0.001, respectively). The subset of triple-negative breast cancers (in other words, those with negative ER, PR, and HER-2/neu status) was positively associated with the methylation of the RIL/CDH13 panel and negatively associated with the HIN-1/RASSF1A panel. Mutations of p53 were found in nine breast tumors (11%), seven of which lacked methylation in both panels. Conclusion We have defined two panels (HIN-1/RASSFIA, and RIL/CDH13) of methylation profiles, which correlated, either positively or negatively, to HR status.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- CpG Islands
- DNA Methylation
- DNA, Neoplasm/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor/physiology
- Humans
- Lymphatic Metastasis/pathology
- Middle Aged
- Promoter Regions, Genetic
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
Collapse
Affiliation(s)
- Weiwei Feng
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Lanlan Shen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Sijin Wen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Daniel G Rosen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jaroslav Jelinek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Xin Hu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Shaoyi Huan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Miao Huang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Kelly K Hunt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Robert C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jean-Pierre J Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Yinhua Yu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
189
|
Hondermarck H, Tastet C, El Yazidi-Belkoura I, Toillon RA, Le Bourhis X. Proteomics of Breast Cancer: The Quest for Markers and Therapeutic Targets. J Proteome Res 2008; 7:1403-11. [DOI: 10.1021/pr700870c] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hubert Hondermarck
- INSERM U 908 (JE-2488) “Signalisation des facteurs de croissance dans le cancer du sein. Protéomique fonctionnelle”, IFR-147, Institut National de la Santé et de la Recherche Médicale and Université Lille 1, France
| | - Christophe Tastet
- INSERM U 908 (JE-2488) “Signalisation des facteurs de croissance dans le cancer du sein. Protéomique fonctionnelle”, IFR-147, Institut National de la Santé et de la Recherche Médicale and Université Lille 1, France
| | - Ikram El Yazidi-Belkoura
- INSERM U 908 (JE-2488) “Signalisation des facteurs de croissance dans le cancer du sein. Protéomique fonctionnelle”, IFR-147, Institut National de la Santé et de la Recherche Médicale and Université Lille 1, France
| | - Robert-Alain Toillon
- INSERM U 908 (JE-2488) “Signalisation des facteurs de croissance dans le cancer du sein. Protéomique fonctionnelle”, IFR-147, Institut National de la Santé et de la Recherche Médicale and Université Lille 1, France
| | - Xuefen Le Bourhis
- INSERM U 908 (JE-2488) “Signalisation des facteurs de croissance dans le cancer du sein. Protéomique fonctionnelle”, IFR-147, Institut National de la Santé et de la Recherche Médicale and Université Lille 1, France
| |
Collapse
|
190
|
Losi-Guembarovski R, Kuasne H, Guembarovski AL, Rainho CA, Cólus IMS. DNA methylation patterns of the CDH1, RARB, and SFN genes in choroid plexus tumors. ACTA ACUST UNITED AC 2008; 179:140-5. [PMID: 18036402 DOI: 10.1016/j.cancergencyto.2007.05.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Accepted: 05/09/2007] [Indexed: 11/17/2022]
Abstract
Genetic and epigenetic alterations in choroid plexus tumors, a rare neuroepithelial neoplasm most frequently detected in children, are poorly characterized. Epigenetic silencing associated with aberrant CpG island methylation is one mechanism leading to the loss of tumor suppressor functions in cancer cells. Using methylation-specific polymerase chain reaction, the methylation patterns of the genes CDH1 (E-cadherin), RARB (retinoic acid receptor, beta), and SFN (stratifin; 14-3-3sigma) were retrospectively investigated in eight choroid plexus tumors (five papillomas, two atypical papillomas, and one carcinoma), as well as in two normal cortexes obtained after autopsy from male individuals aged 6 months and 64 years. Among the six pediatric tumors, the mean age at diagnosis was 1.8 years old (range, 0.2-6) and the two adult tumors were detected in a 66-year-old man and a 45-year-old woman. A high frequency of hypermethylation was detected in CDH1 and SFN genes in tumoral and normal cortex tissues. Tumor-specific RARB hypermethylation was observed in four papillomas. Further studies are required to evaluate the role of aberrant methylation in choroid plexus tumor progression.
Collapse
Affiliation(s)
- Roberta Losi-Guembarovski
- Department of General Biology, Biological Sciences Center, State University of Londrina-UEL, Londrina, PR, Brazil
| | | | | | | | | |
Collapse
|
191
|
Roma AA, Goldblum JR, Fazio V, Yang B. Expression of 14-3-3sigma, p16 and p53 proteins in anal squamous intraepithelial neoplasm and squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2008; 1:419-425. [PMID: 18787619 PMCID: PMC2480570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Accepted: 01/04/2008] [Indexed: 05/26/2023]
Abstract
14-3-3sigma is a p53-regulated G2/M inhibitor involved in numerous cellular signaling pathways related to cell cycle, DNA repair and apoptosis. Recent studies have showed that 14-3-3sigma was silenced transcriptionally through promoter hypermethylation mainly in HPV-negative vulvar squamous cell carcinoma (SCC). However, the expression of 14-3-3sigma protein has not yet been studied in anal SCC and its precursor, anal intraepithelial neoplasm (AIN). In this study, we evaluated the expression of 14-3-3sigma, p16 and p53 in 34 cases of normal perianal squamous mucosa, 5 cases of squamous hyperplasia and 62 cases of AIN, including 54 bowenoid and 8 differentiated AINs. Fourteen cases of invasive anal SCC were also included in the study, including 8 cases associated with bowenoid AIN and 6 cases associated with differentiated AIN. Expression of p16, p53 and 14-3-3sigma proteins was not seen in normal squamous epithelium. Weak staining for 14-3-3sigma was seen in anal squamous hyperplasia. Strong and diffuse p16 immunoreactivity was seen in 98.1% of bowenoid AIN, but only in 12.5% of differentiated AIN. In contrast, increased basal staining with suprabasal extension of p53 was seen in 100% of differentiated AIN, but in none of the bowenoid AIN. Expression of p16 and p53 was essentially mutually exclusive except in one case. Overexpression of 14-3-3sigma was detected in 97% (60/62) of AIN cases, including 96.3% of bowenoid AIN and 100% of differentiated AIN. Expression of 14-3-3sigma was independent of immunoreactivity status for p16 and p53. In conclusion, two histopathologic types of AIN, bowenoid and differentiated, have distinct immunoprofiles for p16 and p53, which suggests dual molecular pathways during anal carcinogenesis. Increased expression of 14-3-3sigma protein was found in approximately 97% of AIN lesions, regardless of histopathologic type and independent of p16 and p53 expression. Our study indicates that immunohistochemical detection of 14-3-3sigma in conjunction with p16 and p53 may be useful in histopathologic recognization of AIN.
Collapse
Affiliation(s)
- Andres A. Roma
- Department of Anatomic Pathology, Division of Surgery, Cleveland Clinic FoundationCleveland, Ohio
| | - John R. Goldblum
- Department of Anatomic Pathology, Division of Surgery, Cleveland Clinic FoundationCleveland, Ohio
| | - Victor Fazio
- Department of Colorectal Surgery, Division of Surgery, Cleveland Clinic FoundationCleveland, Ohio
| | - Bin Yang
- Department of Anatomic Pathology, Division of Surgery, Cleveland Clinic FoundationCleveland, Ohio
| |
Collapse
|
192
|
Down-regulation of 14-3-3zeta suppresses anchorage-independent growth of lung cancer cells through anoikis activation. Proc Natl Acad Sci U S A 2007; 105:162-7. [PMID: 18162532 DOI: 10.1073/pnas.0710905105] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The family of 14-3-3 proteins has emerged as critical regulators of diverse cellular responses under both physiological and pathological conditions. Here, we report an important role of 14-3-3zeta in tumorigenesis through a mechanism that involves anoikis resistance. 14-3-3zeta is up-regulated in a number of cancer types, including lung cancer. Through an RNAi approach using human lung adenocarcinoma-derived A549 cells as a model system, we have found that knockdown of a single zeta isoform of 14-3-3 is sufficient to restore the sensitivity of cancer cells to anoikis and impair their anchorage-independent growth. Enhanced anoikis appears to be mediated in part by up-regulated BH3-only proteins, Bad and Bim, coupled with decreased Mcl-1, resulting in the subsequent activation of Bax. This study suggests a model in which anchorage-independent growth of lung cancer cells requires the presence of 14-3-3zeta. This work not only reveals a critical role of 14-3-3zeta in anoikis suppression in lung cancer cells, but also identifies and validates 14-3-3zeta as a potential molecular target for anticancer therapeutic development.
Collapse
|
193
|
Motokura T, Nakamura Y, Sato H. Aberrant overexpression of an epithelial marker, 14-3-3sigma, in a subset of hematological malignancies. BMC Cancer 2007; 7:217. [PMID: 18036248 PMCID: PMC2222637 DOI: 10.1186/1471-2407-7-217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 11/25/2007] [Indexed: 01/25/2023] Open
Abstract
Background 14-3-3σ is a p53-mediated cell-cycle inhibitor in epithelial cells. The expression of 14-3-3σ is frequently altered in cancers of epithelial origin associated with altered DNA methylation. Since its involvement in a non-epithelial tumor is unknown, we examined 14-3-3σ expression in patients with haematological malignancies. Methods We analyzed 41 hematopoietic cell lines and 129 patients with a variety of hematological malignancies for 14-3-3σ expression with real-time RT-PCR. We also examined protein levels by Western blot analysis and DNA methylation status of the 14-3-3σ gene by methylation-specific PCR analysis of bisulfite-treated DNA. In addition, mutations of p53 gene were identified by RT-PCR-SSCP analysis and the expression levels of 14-3-3σ were compared with those of other cell-cycle inhibitor genes, CDKN2A and ARF. Results The expression levels of 14-3-3σ mRNA in almost all cell lines were low and comparable to those in normal hematopoietic cells except for 2 B-cell lines. On the contrary, 14-3-3σ mRNA was aberrantly overexpressed frequently in mature lymphoid malignancies (30 of 93, 32.3%) and rarely in acute leukemia (3 of 35, 8.6%). 14-3-3σ protein was readily detectable and roughly reflected the mRNA level. In contrast to epithelial tumors, methylation status of the 14-3-3σ gene was not associated with expression in hematological malignancies. Mutations of p53 were identified in 12 patients and associated with lower expression of 14-3-3σ. The expression levels of 14-3-3σ, CDKN2A and ARF were not correlated with but rather reciprocal to one another, suggesting that simultaneous overexpression of any two of them is incompatible with tumor growth. Conclusion 14-3-3σ, an epithelial cell marker, was overexpressed significantly in a subset of mature lymphoid malignancies. This is the first report of aberrant 14-3-3σ expression in non-epithelial tumors in vivo. Since the significance of 14-3-3σ overexpression is unknown even in epithelial tumors such as pancreatic cancers, further analysis of regulation and function of the 14-3-3σ gene in non-epithelial as well as epithelial tumors is warranted.
Collapse
Affiliation(s)
- Toru Motokura
- Department of Hematology and Oncology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| | | | | |
Collapse
|
194
|
Ai L, Kim WJ, Kim TY, Fields CR, Massoll NA, Robertson KD, Brown KD. Epigenetic silencing of the tumor suppressor cystatin M occurs during breast cancer progression. Cancer Res 2007; 66:7899-909. [PMID: 16912163 DOI: 10.1158/0008-5472.can-06-0576] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cystatin M is a secreted inhibitor of lysosomal cysteine proteases. Several lines of evidence indicate that cystatin M is a tumor suppressor important in breast malignancy; however, the mechanism(s) that leads to inactivation of cystatin M during cancer progression is unknown. Inspection of the human cystatin M locus uncovered a large and dense CpG island within the 5' region of this gene (termed CST6). Analysis of cultured human breast tumor lines indicated that cystatin M expression is either undetectable or in low abundance in several lines; however, enhanced gene expression was measured in cells cultured on the DNA demethylating agent 5-aza-2'-deoxycytidine (5-aza-dC). Increased cystatin M expression does not correlate with a cytotoxic response to 5-aza-dC; rather, various molecular approaches indicated that the CST6 gene was aberrantly methylated in these tumor lines as well as in primary breast tumors. Moreover, 60% (12 of 20) of primary tumors analyzed displayed CST6 hypermethylation, indicating that this aberrant characteristic is common in breast malignancies. Finally, preinvasive and invasive breast tumor cells were microdissected from nine archival breast cancer specimens. Of the five tumors displaying CST6 gene methylation, four tumors displayed methylation in both ductal carcinoma in situ and invasive breast carcinoma lesions and reduced expression of cystatin M in these tumors was confirmed by immunohistochemistry. In summary, this study establishes that the tumor suppressor cystatin M is a novel target for epigenetic silencing during mammary tumorigenesis and that this aberrant event can occur before development of invasive breast cancer.
Collapse
Affiliation(s)
- Lingbao Ai
- Department of Biochemistry and Molecular Biology and University of Florida Shands Cancer Center Program in Cancer Genetics, Epigenetics, and Tumor Virology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | | | |
Collapse
|
195
|
Abstract
Genetic and epigenetic mechanisms contribute to the development of human tumors. However, the conventional analysis of neoplasias has preferentially focused on only one of these processes. This approach has led to a biased, primarily genetic view, of human tumorigenesis. Epigenetic alterations, such as aberrant DNA methylation, are sufficient to induce tumor formation, and can modify the incidence, and determine the type of tumor which will arise in genetic models of cancer. These observations raise important questions about the degree to which genetic and epigenetic mechanisms cooperate in human tumorigenesis, the identity of the specific cooperating genes and how these genes interact functionally to determine the diverse biological and clinical paths to tumor initiation and progression. These gaps in our knowledge are, in part, due to the lack of methods for full-scale integrated genetic and epigenetic analyses. The ultimate goal to fill these gaps would include sequencing relevant regions of the 3-billion nucleotide genome, and determining the methylation status of the 28-million CpG dinucleotide methylome at single nucleotide resolution in different types of neoplasias. Here, we review the emergence and advancement of technologies to map ever larger proportions of the cancer methylome, and the unique discovery potential of integrating these with cancer genomic data. We discuss the knowledge gained from these large-scale analyses in the context of gene discovery, therapeutic application and building a more widely applicable mechanism-based model of human tumorigenesis.
Collapse
Affiliation(s)
- Romulo M Brena
- Department of Molecular Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | | |
Collapse
|
196
|
Zhu F, Xia X, Liu B, Shen J, Hu Y, Person M, Hu Y. IKKalpha shields 14-3-3sigma, a G(2)/M cell cycle checkpoint gene, from hypermethylation, preventing its silencing. Mol Cell 2007; 27:214-227. [PMID: 17643371 DOI: 10.1016/j.molcel.2007.05.042] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 01/22/2007] [Accepted: 05/25/2007] [Indexed: 11/27/2022]
Abstract
We recently reported that a large proportion of aggressive squamous cell carcinomas of humans and mice express markedly reduced IKKalpha. However, the role of IKKalpha in maintaining genomic stability is unknown. Here we reported that IKKalpha-deficient keratinocytes had a defect in the G(2)/M cell-cycle arrest in response to DNA damage due to downregulated 14-3-3sigma, a cell cycle checkpoint protein. Trimethylated histone H3 lysine 9 (H3-K9) was found to associate with the histone trimethyltransferase Suv39h1 and DNA methyltransferase Dnmt3a in the methylated 14-3-3sigma locus. Reintroduction of IKKalpha restored the expression of 14-3-3sigma. IKKalpha was found to associate with H3 in 14-3-3sigma, which prevented access of Suv39h1 to H3, thereby preventing hypermethylation of 14-3-3sigma. IKKalpha mutants that failed to bind to H3 did not restore the expression of 14-3-3sigma. Thus, IKKalpha protects the 14-3-3sigma locus from hypermethylation, which serves as a mechanism of maintaining genomic stability in keratinocytes.
Collapse
Affiliation(s)
- Feng Zhu
- Science Park Research Division, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Xiaojun Xia
- Science Park Research Division, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Bigang Liu
- Science Park Research Division, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Jianjun Shen
- Science Park Research Division, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Yuhui Hu
- Science Park Research Division, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | - Maria Person
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yinling Hu
- Science Park Research Division, The University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA.
| |
Collapse
|
197
|
Sigalotti L, Fratta E, Coral S, Cortini E, Covre A, Nicolay HJM, Anzalone L, Pezzani L, Di Giacomo AM, Fonsatti E, Colizzi F, Altomonte M, Calabrò L, Maio M. Epigenetic drugs as pleiotropic agents in cancer treatment: biomolecular aspects and clinical applications. J Cell Physiol 2007; 212:330-44. [PMID: 17458893 DOI: 10.1002/jcp.21066] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In the last three decades huge efforts have been made to characterize genetic defects responsible for cancer development and progression, leading to the comprehensive identification of distinct cellular pathways affected by the alteration of specific genes. Despite the undoubtable role of genetic mechanisms in triggering neoplastic cell transformation, epigenetic modifications (i.e., heritable changes of gene expression that do not derive from alterations of the nucleotide sequence of DNA) are rapidly emerging as frequent alterations that often occur in the early phases of tumorigenesis and that play an important role in tumor development and progression. Epigenetic alterations, such as modifications in DNA methylation patterns and post-translational modifications of histone tails, behave extremely different from genetic modifications, being readily revertable by "epigenetic drugs" such as inhibitors of DNA methyl transferases and inhibitors of histone deacetylases. Since epigenetic alterations in cancer cells affect virtually all cellular pathways that have been associated to tumorigenesis, it is not surprising that epigenetic drugs display pleiotropic activities, being able to concomitantly restore the defective expression of genes involved in cell cycle control, apoptosis, cell signaling, tumor cell invasion and metastasis, angiogenesis and immune recognition. Prompted by this emerging clinical relevance of epigenetic drugs, this review will focus on the large amount of available data, deriving both from in vitro experimentations and in vivo pre-clinical and clinical studies, which clearly indicate epigenetic drugs as effective modifiers of cancer phenotype and as positive regulators of tumor cell biology with a relevant therapeutic potential in cancer patients.
Collapse
Affiliation(s)
- Luca Sigalotti
- Cancer Bioimmunotherapy Unit, Department of Medical Oncology, Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Kuznetsova EB, Kekeeva TV, Larin SS, Zemlyakova VV, Babenko OV, Nemtsova MV, Zaletayev DV, Strelnikov VV. Novel markers of gene methylation and expression in breast cancer. Mol Biol 2007. [DOI: 10.1134/s0026893307040061] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
199
|
Medina A, Ghaffari A, Kilani RT, Ghahary A. The role of stratifin in fibroblast-keratinocyte interaction. Mol Cell Biochem 2007; 305:255-64. [PMID: 17646930 DOI: 10.1007/s11010-007-9538-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 06/21/2007] [Indexed: 10/23/2022]
Abstract
Stratifin is a member of 14-3-3 protein family, a highly conserved group of proteins constituted by seven isoforms. They are involved in numerous crucial intracellular functions such as cell cycle and apoptosis, regulation of signal transduction pathways, cellular trafficking, cell proliferation and differentiation, cell survival, and protein folding and processing, among others. At epidermal level, stratifin (also called 14-3-3 sigma) has been described as molecule with relevant functions. For instance, this isoform is a marker associated with keratinocyte differentiation. In this maturation process, the presence of dominant negative molecules of p53 induces a "stemness condition" of keratinocyte precursor cells and suppression of stratifin expression. In addition, the recently described keratinocyte-releasable form of stratifin is involved in dermal fibroblast MMP-1 over-expression through c-Fos and c-Jun activity. This effect is mediated, at least in part, by p38 mitogen-activated protein kinase (MAPK). Other MMP family members such as stromelysin-1 (MMP-3), stromelysin-2 (MMP-10), neutrophil collagenase (MMP-8), and membrane-type MMP-24 (MT5-MMP) are also up-regulated by stratifin. Within fibroproliferative disorder of skin, hypertrophic scar and keloids exhibit a high content of collagen, proteoglycans, and fibronectin. Thus, the MMP profile induced by stratifin is an interesting starting point to establish new therapeutic tools to control the process of wound healing. In this review, we will focus on site of synthesis and mode of action of stratifin in skin and wound healing.
Collapse
Affiliation(s)
- Abelardo Medina
- BC Professional Fire Fighters' Burn and Wound Healing Laboratory, Division of Plastic Surgery, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
200
|
Jeronimo C, Monteiro P, Henrique R, Dinis-Ribeiro M, Costa I, Costa VL, Filipe L, Carvalho AL, Hoque MO, Pais I, Leal C, Teixeira MR, Sidransky D. Quantitative hypermethylation of a small panel of genes augments the diagnostic accuracy in fine-needle aspirate washings of breast lesions. Breast Cancer Res Treat 2007; 109:27-34. [PMID: 17549626 DOI: 10.1007/s10549-007-9620-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2007] [Accepted: 05/09/2007] [Indexed: 12/31/2022]
Abstract
PURPOSE We hypothesized that comprehensive breast cancer methylation profiling might provide biomarkers for diagnostic assessment of suspicious breast lesions using fine needle aspiration biopsy (FNA). EXPERIMENTAL DESIGN Twenty-three gene promoters were surveyed by quantitative methylation-specific PCR in bisulfite-modified DNA from 66 breast carcinomas (BCa), 31 fibroadenomas (FB) and 12 normal breast (NT) samples to define a set of genes differentially methylated in malignant and non-malignant tissues. This set was tested in 78 FNA washings obtained pre-operatively (66 malignant, 12 benign), with histopathological diagnosis. Receiver operator characteristic (ROC) curve analysis identified a gene panel which might distinguish cancer from non-cancerous lesions. Finally, this panel was validated in an independent series of FNA washings (45 cases) in which cytomorphology did not reach definitive diagnosis. RESULTS In tissue samples, 14-3-3-sigma, DAPK, CCND2, RASSF1A, CALCA, APC, HIN1, RARbeta2, TIG1, and GSTP1 methylation levels differed significantly among BCa, FB, and NT. ROC curve analysis identified a panel of four gene loci (CCND2, RASSF1A, APC, and HIN1) that discriminated BCa from benign lesions in a set of 78 FNA washings from histologically characterized breast lesions. When this panel was tested in the validation dataset of 45 FNA washings, breast cancer was identified with perfect specificity (100%) when 3 of 4 gene loci tested positive, providing estimated added information of 91% over cytomorphologic evaluation alone. CONCLUSIONS Our data provide evidence that multigene methylation analysis augments diagnostic accuracy of cytological assessment of suspicious breast lesions, and might be a valuable ancillary tool for breast cancer diagnosis.
Collapse
Affiliation(s)
- Carmen Jeronimo
- Department of Genetics, Portuguese Oncology Institute, Rua Dr. Antonio Bernardino Almeida, Porto, Portugal.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|